US7794977B2 - Means and methods for regulating gene expression - Google Patents

Means and methods for regulating gene expression Download PDF

Info

Publication number
US7794977B2
US7794977B2 US11/888,568 US88856807A US7794977B2 US 7794977 B2 US7794977 B2 US 7794977B2 US 88856807 A US88856807 A US 88856807A US 7794977 B2 US7794977 B2 US 7794977B2
Authority
US
United States
Prior art keywords
star
sequence
cell
seq
cell line
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related, expires
Application number
US11/888,568
Other versions
US20080131930A1 (en
Inventor
Arie P. Otte
Arthur L. Kruckeberg
David P. E. Satijn
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Chromagenics BV
Original Assignee
Chromagenics BV
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Chromagenics BV filed Critical Chromagenics BV
Priority to US11/888,568 priority Critical patent/US7794977B2/en
Assigned to CHROMAGENICS B.V. reassignment CHROMAGENICS B.V. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SATIJN, DAVID P. E., OTTE, ARIE P., KRUCKEBERG, ARTHUR L.
Priority to US11/978,483 priority patent/US20080206813A1/en
Publication of US20080131930A1 publication Critical patent/US20080131930A1/en
Application granted granted Critical
Publication of US7794977B2 publication Critical patent/US7794977B2/en
Adjusted expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/67General methods for enhancing the expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/02Preparation of peptides or proteins having a known sequence of two or more amino acids, e.g. glutathione
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10322New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/108Plasmid DNA episomal vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/001Vector systems having a special element relevant for transcription controllable enhancer/promoter combination
    • C12N2830/005Vector systems having a special element relevant for transcription controllable enhancer/promoter combination repressible enhancer/promoter combination, e.g. KRAB
    • C12N2830/006Vector systems having a special element relevant for transcription controllable enhancer/promoter combination repressible enhancer/promoter combination, e.g. KRAB tet repressible
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/48Vector systems having a special element relevant for transcription regulating transport or export of RNA, e.g. RRE, PRE, WPRE, CTE
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/80Vector systems having a special element relevant for transcription from vertebrates
    • C12N2830/85Vector systems having a special element relevant for transcription from vertebrates mammalian
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/20Vectors comprising a special translation-regulating system translation of more than one cistron
    • C12N2840/203Vectors comprising a special translation-regulating system translation of more than one cistron having an IRES

Definitions

  • the invention relates to the fields of medicine and cellular biology. More specifically, the invention relates to means and methods for regulating gene expression, and production of proteinaceous molecules.
  • Polypeptide production at industrial scale currently provides many biologically active polypeptides for a variety of uses, including diagnostic and therapeutic pharmaceuticals, industrial catalysts and nourishment.
  • Polypeptides are produced in a variety of host systems, including transgenic animals and plants, microbes, and cultured mammalian cells. In most cases, the host system is modified by recombinant DNA techniques, for instance resulting in introduction into the host cell of a transgene which encodes a polypeptide of interest.
  • a transgene typically includes elements that influence the transcription, translation, and/or processing of the transgene's polypeptide coding sequence.
  • a recombinant host is then identified and isolated which has a suitable yield of a polypeptide of interest, and the cell population of this recombinant host is increased to an extent that it can produce the required amount of polypeptide.
  • the choice of the host system depends on a number of factors including: (1) the nature and intended use of a polypeptide, and (2) the cost of production.
  • the host system of choice is usually cultured mammalian cells. Considerations with respect to product use and production cost with host cells will be discussed below.
  • a therapeutic protein For in vivo therapeutic use, a therapeutic protein must not only have the correct biological activity to alter the course of a disease. It must also do no harm. Most therapeutic proteins are exported from the cell by the secretory pathway. Secreted proteins are modified by a series of post-translational events, including glycosylation, disulfide bond formation, and proteolytic processing. The post-translational modification systems vary among different species and cell types in their detailed mechanisms of action. As a result, the same polypeptide chain can be detectably different when it is produced in different host cells. These differences can be analytical, such as differences in physical properties such as molecular mass, net electrical charge, carbohydrate composition, or structure.
  • the differences can also be functional, affecting for instance the biological activity of the protein itself (catalytic activity, ligand binding characteristics, etc.), and/or its in vivo properties (immunogenicity, biological half life, biodistribution, etc.). Functional differences can, therefore, affect both function and possible side effect(s) of a therapeutic protein.
  • Host cell lines that produce proteins with low efficacy are not suitable for commercial exploitation.
  • host cells which produce modified protein that involves significant side effects in a patient should not be used. These factors are becoming increasingly important considerations during selection of a host cell line for production of a therapeutic protein.
  • Therapeutic protein production in host cells is an intrinsically costly process. Current methods for industrial production of such proteins often perform poorly, resulting in products that are prohibitively expensive. Poor performance can be due to limitations of protein expression systems and host cell lines currently in use. These limitations mostly have a few specific causes, including (a) failure to identify and isolate recombinant host cell lines that have suitable productivity of proteins (poor predictability), (b) silencing, during the industrial production cycle, of the transgenes that encode proteins (poor stability), and (c) low or incorrect post-translational processing and secretory capacity of the host cell line. These limitations will be considered separately below.
  • Transgenes are often subject to silencing during cultivation of recombinant host cells. Silencing acts by suppressing transcription of a transgene. Detailed mechanisms of silencing are still not known, and different conventional methods are prone to different kinds of silencing phenomena. With one phenomenon, an individual transgene is silenced by formation of transcriptionally refractory heterochromatin at the transgenic locus. Heterochromatin formation is influenced by the position of genomic integration of a transgene (“position effects” (Boivin and Dura, 1998)). Transgene integration occurs more or less at random. Since most of the genome is heterochromatin, most transgene loci are prone to silencing due to position effects.
  • a second transgene-silencing phenomenon can occur when two or more copies of a transgene are integrated into a genome during construction of a recombinant cell line. Formation of tandem transgene repeats often occurs during the initial integration step. Furthermore, in order to increase product yield, many recombinant host cell lines are engineered after the integration step to amplify the copy number of a transgene, which also results in tandem transgene repeats (Kaufman, 1990). Tandem repeats and other configurations of multiple transgene copies are particularly prone to silencing (“repeat-induced gene silencing” (Garrick et al., 1998)).
  • the yield can also decline via another phenomenon than transcriptional silencing.
  • the number of copies of the transgene can decline during cultivation of a recombinant host cell line.
  • the productivity of such cell lines at the time of selection for use is correlated with a transgene copy number, and consequently as copies of a transgene are lost, the yield declines (Kaufman, 1990).
  • Different cell types in a mammalian organism have different capacities for post-translational modification and secretion of proteins.
  • the functions of some cell types include production of large quantities of secreted proteins; examples include lymphocytes (producing immunoglobulins), hepatocytes (producing serum proteins), and fibroblasts (producing extracellular matrix proteins).
  • lymphocytes producing immunoglobulins
  • hepatocytes producing serum proteins
  • fibroblasts producing extracellular matrix proteins.
  • These cell types are favorable sources for deriving host cell lines for production of secreted heterologous proteins. More favorable is the use of a cell line whose progenitor organismal cell type secretes a protein or class of proteins of interest.
  • lymphocytes or host cells derived from lymphocytes
  • erythropoietin in hepatocytes
  • host cells derived from hepatocytes or host cells derived from hepatocytes
  • blood clotting factors e.g., Factor VIII and van Willebrand's factor
  • heterologous neurotrophins with post-translational modifications (and hence functional properties) that match the naturally-occurring proteins
  • expression in host cells derived from the central nervous system is favorable.
  • production of polypeptides such as neurotrophins in host cell lines such as those derived from neural tissue is inefficient using conventional methods.
  • the predictability of identifying high-expressor isolates from these types of cell lines is often poor; the yield of proteins from such cell lines is generally low, and production levels are characteristically unstable.
  • Another drawback to a use of specific host cells for production of affiliated proteins is that it is usually difficult to isolate cell lines with favorable biotechnological characteristics. These characteristics for instance include the mode and rate of growth, and the ease of introduction of a transgene. Consequently, various general host cell lines have been established. Examples of these include CHO cells from Chinese hamster ovary (ATCC (American Type Culture Collection) CCL-61), BHK cells from baby hamster kidney (ATCC CCL-10), and Vero cells from African green monkey kidney (ATCC CCL-81). These “general purpose” host cell lines are widely used for production of a number of heterologous proteins. A disadvantage of general purpose cell lines is that the post-translational modifications of heterologous proteins produced by them often differ from the post-translational modifications of the naturally occurring proteins. These differences can have functional consequences resulting in side effects, as discussed above.
  • Table 1 lists a number of proteins that are currently in use or under development for biopharmaceutical applications. It also lists the tissue or cell type in which each protein is normally produced in the human body. These 24 proteins (or protein classes) come from a wide range of cells and tissue, ranging from highly secretory cells (hepatocytes, endothelial cells) to cells with low secretory capacity (e.g., neural tissue). Currently, neither general-purpose host cells nor specific host cells have qualities that enable optimal expression of the broad spectrum of biopharmaceutically important secreted proteins.
  • the present invention provides a method for producing a proteinaceous molecule in a cell comprising selecting a cell for its suitability for producing the proteinaceous molecule, providing a nucleic acid encoding the proteinaceous molecule with a nucleic acid comprising a STAR (STabilizing Anti-Repression) sequence, expressing the resulting nucleic acid in the cell and collecting the proteinaceous molecule.
  • a STAR STabilizing Anti-Repression
  • the STAR sequence has to be operably linked to the nucleic acid encoding the proteinaceous molecule in order to be effective.
  • one STAR element is used.
  • more than one STAR element is used.
  • the nucleic acid encoding the proteinaceous molecule is provided with two STAR sequences; one STAR sequence at the 5′ side of the coding sequence of the nucleic acid and one STAR sequence at the 3′ side of the coding sequence of the nucleic acid.
  • STAR sequences are nucleic acid sequences that comprise a capacity to influence transcription of genes in cis. Typically, although not necessarily, the STAR sequences do not code by themselves for a functional protein.
  • a STAR sequence has a gene transcription modulating quality in at least one type of cell.
  • a STAR sequence is capable of enhancing gene transcription resulting in a higher yield, increasing the proportion of transgene-comprising host cells with acceptable expression levels, and/or increasing stability of transgenes in recombinant cell lines.
  • a method of detecting, and optionally selecting, a STAR sequence comprising providing a transcription system with a variety of a fragment-comprising vectors, the vectors comprising i) an element with a gene-transcription repressing quality, and ii) a promoter directing transcription of a reporter gene, the method further comprising performing a selection step in the transcription system in order to identify the STAR sequence.
  • the fragments are located between i) the element with a gene-transcription repressing quality, and ii) the promoter directing transcription of the reporter gene.
  • RNA polymerase initiates the transcription process after binding to a specific sequence, called the promoter, that signals where RNA synthesis should begin.
  • a STAR sequence can enhance transcription from the promoter in cis, in a given cell type and/or a given promoter.
  • STAR elements have at least one of three consequences for production of (heterologous) proteinaceous molecule (also referred to as (heterologous) protein): (1) they increase the predictability of identifying host cell lines that express a proteinaceous molecule at industrially acceptable levels; (2) they result in host cell lines with increased protein yields; and/or (3) they result in host cell lines that exhibit more stable protein production during prolonged cultivation.
  • (heterologous) protein also referred to as (heterologous) protein
  • transgene expression units can occur at random positions throughout the host cell genome. However, much of the genome is transcriptionally silent heterochromatin. When the expression units include STAR elements flanking the transgene, the position of integration has a reduced effect on expression. The STAR elements impair the ability of adjacent heterochromatin to silence the transgene. Consequently, the proportion of transgene-containing host cells with acceptable expression levels is increased.
  • STAR elements increase the stability of transgenes in recombinant host cell lines by ensuring that the transgenes are not transcriptionally silenced during prolonged cultivation. Comparative trials show that, under conditions in which transgenes that are not protected by STAR elements are progressively silenced (5 to 25 passages in cultivation), STAR element-protected transgenes continue to be expressed at high levels. This is an advantage during industrial production of proteinaceous molecules, during which cell cultivation continues for prolonged periods, from a few weeks to many months.
  • a STAR sequence can enhance expression of a heterologous proteinaceous molecule.
  • a STAR sequence can enhance expression of a naturally produced proteinaceous molecule.
  • Transcription can be influenced through a direct effect of the STAR sequence (or the protein(s) binding to it) on the transcription of a particular promoter. Transcription can however, also be influenced by an indirect effect, for instance because the STAR sequence affects the function of one or more other regulatory elements.
  • a STAR sequence can also comprise a stable gene transcription quality. Frequently, expression levels drop dramatically with increasing numbers of cell divisions. With the methods disclosed in EP 01202581.3 it is possible to detect and optionally select a DNA sequence that is capable of at least in part preventing the dramatic drop in transcription levels with increasing numbers of cell divisions. Strikingly, fragments comprising a STAR sequence can be detected and optionally selected with a method EP 01202581.3, in spite of the fact that the method does not necessarily measure long term stability of transcription.
  • a STAR sequence is suitable for enhancing the level of transcription of a gene of interest in a host cell. If, together with a gene of interest, a STAR sequence is also introduced into host cells, more clones can be detected that express more than a certain amount of the gene of interest. As used herein, such host cells are termed “host cells with acceptable expression levels.”
  • STAR sequence derived from a plant and/or vertebrate is used. More preferably a human STAR sequence is used.
  • Sequences comprising a STAR sequence can be found by using a functional assay, as described above. However, once a collection of such sequences has been identified, bioinformatics can be used to find other STAR sequences.
  • bioinformatics can be used to find other STAR sequences.
  • sequence identifiers can be extracted from a family of DNA sequences sharing a certain common feature. Such sequence identifiers can subsequently be used to identify sequences that share one or more identifiers. Sequences sharing such one or more identifiers are likely to be a member of the same family of sequences, i.e., are likely to share the common feature of the family.
  • STAR sequences sequence identifiers
  • patterns can be used to determine whether a test sequence is likely to contain STAR activity.
  • a method for detecting the presence of a STAR sequence within a nucleic acid sequence of about 50-5000 base pairs comprising determining the frequency of occurrence in the sequence of at least one sequence pattern and determining that the frequency of occurrence is representative of the frequency of occurrence of at least one sequence pattern in at least one sequence comprising a STAR sequence.
  • any method is suited for determining whether a sequence pattern is representative of a STAR sequence. Many different methods are available in the art.
  • the step of determining that the occurrence is representative of the frequency of occurrence of at least one sequence pattern in at least one sequence comprising a STAR sequence comprises, determining that the frequency of occurrence of at least one sequence pattern significantly differs between at least one STAR sequence and at least one control sequence. In principle any significant difference is discriminative for the presence of a STAR sequence. However, in a particularly preferred embodiment, the frequency of occurrence of at least one sequence pattern is significantly higher in at least one sequence comprising a STAR sequence compared to at least one control sequence.
  • sequences comprising a STAR sequence have been identified by the present inventors. It is possible to use these sequences to test how efficient a pattern is in discriminating between a control sequence and a sequence comprising a STAR sequence. Using so-called discriminant analysis it is possible to determine on the basis of any set of STAR sequences in a species, the most optimal discriminative sequence patterns or combination thereof. Thus, preferably, at least one of the patterns is selected on the basis of optimal discrimination between at least one sequence comprising a STAR sequence and a control sequence.
  • the frequency of occurrence of a sequence pattern in a test nucleic acid is compared with the frequency of occurrence in a sequence known to contain a STAR sequence.
  • a pattern is considered representative for a sequence comprising a STAR sequence if the frequencies of occurrence are similar.
  • another criterion is used.
  • the frequency of occurrence of a pattern in a sequence comprising a STAR sequence is compared to the frequency of occurrence of the pattern in a control sequence.
  • a sequence pattern is considered to be representative of a sequence comprising a STAR sequence, if the frequency of occurrence of the pattern in at least one sequence comprising a STAR sequence is significantly different from the frequency of occurrence of the same pattern in a control sequence.
  • the frequency of occurrence is representative of the frequency of occurrence of at least one sequence pattern in at least two sequences comprising a STAR sequence; more preferably, in at least five sequences comprising a STAR sequence; and, even more preferably, in at least ten sequences comprising a STAR sequence. More preferably, the frequency of occurrence is representative of the frequency of occurrence of at least one sequence pattern in at least 20 sequences comprising a STAR sequence. In a particularly preferred embodiment, the frequency of occurrence is representative of the frequency of occurrence of at least one sequence pattern in at least 50 sequences comprising a STAR.
  • the patterns that are indicative for a sequence comprising a STAR sequence are also dependent on the type of control nucleic acid used.
  • the type of control sequence used is preferably selected on the basis of the sequence in which the presence of a STAR sequence is to be detected.
  • the control sequence comprises a random sequence comprising a similar AT/CG content as at least one sequence comprising a STAR sequence.
  • the control sequence is derived from the same species as the sequence comprising the STAR sequence. For instance, if a test sequence is scrutinized for the presence of a STAR sequence, active in a plant cell, then preferably the control sequence is also derived from a plant cell.
  • control nucleic acid is preferably also derived from a human genome.
  • control sequence comprises between 50% and 150% of the bases of at least one sequence comprising a STAR sequence.
  • control sequence comprises between 90% and 110% of the bases of at least one sequence comprising a STAR sequence. More preferably, between 95% and 105%.
  • a pattern can comprise any number of bases larger than two.
  • at least one sequence pattern comprises at least five, more preferably at least six, bases.
  • at least one sequence pattern comprises at least eight bases.
  • the at least one sequence pattern comprises a pattern listed in Table 6 and/or Table 7.
  • a pattern may consist of a consecutive list of bases. However, the pattern may also comprise bases that are interrupted one or more times by a number of bases that are not or only partly discriminative. A partly discriminative base is, for instance, indicated as a purine.
  • the presence of STAR activity is verified using a functional assay.
  • a sequence comprises STAR activity.
  • STAR activity is confirmed if the sequence is capable of performing at least one of the following functions: (i) at least in part inhibiting the effect of sequence comprising a gene transcription repressing element of the invention, (ii) at least in part blocking chromatin-associated repression, (iii) at least in part blocking activity of an enhancer, (iv) conferring upon an operably linked nucleic acid encoding a transcription unit compared to the same nucleic acid alone, (iv-a) a higher predictability of transcription, (iv-b) a higher transcription, and/or (iv-c) a higher stability of transcription over time.
  • sequences comprising STAR activity identified by the present inventors open up a wide variety of possibilities to generate and identify sequences comprising the same activity in kind not necessarily in amount. For instance, it is well within the reach of a skilled person to alter the sequences identified in the present invention and test the altered sequence for STAR activity. Such altered sequences are, therefore, also part of the present invention. Alteration can include deletion, insertion and mutation of one or more bases in the sequences.
  • Sequences comprising STAR activity were identified in stretches of 400 bases. However, it is expected that not all of these 400 bases are required to retain STAR activity. Methods to delimit the sequences that confer a certain property to a fragment of between 400 and 5000 bases are well known. The minimal sequence length of a fragment comprising STAR activity is estimated to be about 50 bases.
  • Table 6 (SEQ ID NOS:177-342) and Table 7 (SEQ ID NOS:343-1072) list patterns of six bases that have been found to be over represented in nucleic acid molecules comprising STAR activity. This over representation is considered to be representative for a STAR sequence.
  • the tables were generated for a family of 65 STAR sequences (SEQ ID NOS:1-65). Similar tables can be generated starting from a different set of STAR sequences, or from a smaller or larger set of STAR sequences.
  • a pattern is representative for a STAR sequence if it is over represented in the STAR sequence compared to a sequence not comprising a STAR element. This can be a random sequence.
  • the sequence comprising a STAR sequence is preferably compared to a genome or a significant part thereof.
  • a genome of a vertebrate or plant more preferably, a human genome.
  • a significant part of a genome is, for instance, a chromosome.
  • the sequence comprising a STAR sequence and the control sequence are derived from nucleic acid of the same species.
  • the frequency of occurrence is representative of the frequency of occurrence of at least one sequence pattern in at least two sequences comprising a STAR sequence; more preferably, in at least five sequences comprising a STAR sequence; and, even more preferably, in at least ten sequences comprising a STAR sequence.
  • the frequency of occurrence is representative of the frequency of occurrence of at least one sequence pattern in at least 20 sequences comprising a STAR sequence.
  • the frequency of occurrence is representative of the frequency of occurrence of at least one sequence pattern in at least 50 sequences comprising a STAR.
  • the sequences comprising a STAR sequence comprises at least one of the sequences depicted in the sequences comprising STAR1-STAR65 (SEQ ID NOS:1-65), sequences comprising STAR66 and testing set (SEQ ID NOS:66-84), and sequences comprising Arabidopsis STAR A1-A35 (SEQ ID NOS:85-119) (hereinafter SEQ ID NOS:1-119).
  • STAR activity is a feature shared by the sequences listed in SEQ ID NOS:1-119. However, this does not mean that they must all share the same identifier sequence. It is very well possible that different identifiers exist. Identifiers may confer this common feature onto a fragment containing it, though this is not necessarily so.
  • At least one of the sequence patterns for detecting a STAR sequence comprises a sequence pattern GGACCC (SEQ ID NO:464), CCCTGC (SEQ ID NO:816), AAGCCC (SEQ ID NO:270), CCCCCA (SEQ ID NO:298) and/or AGCACC (SEQ ID NO:336).
  • at least one of the sequence patterns for detecting a STAR sequence comprises a sequence pattern CCCN ⁇ 16 ⁇ AGC (SEQ ID NO:415), GGCN ⁇ 9 ⁇ GAC (SEQ ID NO:536), CACN ⁇ 13 ⁇ AGG (SEQ ID NO:761), and/or CTGN ⁇ 4 ⁇ GCC (SEQ ID NO:839).
  • a list of STAR sequences can also be used to determine one or more consensus sequences therein.
  • the invention therefore, also provides a consensus sequence for a STAR element. This consensus sequence can of course be used to identify candidate STAR elements in a test sequence.
  • sequences comprising a STAR element can be used by means of sequence homology to identify sequences comprising a STAR element in other species belonging to vertebrate.
  • a mammalian STAR sequence is used to screen for STAR sequences in other mammalian species.
  • STAR sequence can be used to screen for homologous sequences with similar function in other plant species.
  • the invention in one aspect provides a STAR sequence obtainable by a method according to the invention. Further provided is a collection of STAR sequences.
  • the STAR sequence is a vertebrate or plant STAR sequence.
  • the STAR sequence is a mammalian STAR sequence or an angiosperm (monocot, such as rice or dicot, such as Arabidopsis ). More preferably, the STAR sequence is a primate and/or human STAR sequence.
  • a list of sequences comprising STAR activity can be used to determine whether a test sequence comprises a STAR element.
  • the invention provides a method for determining whether a nucleic acid sequence of about 50-5000 base pairs comprises a STAR sequence, the method comprising: generating a first table of sequence patterns comprising the frequency of occurrence of the patterns in a collection of STAR sequences of the invention; generating a second table of the patterns comprising the frequency of occurrence of the patterns in at least one reference sequence; selecting at least one pattern of which the frequency of occurrence differs between the two tables; determining, within the nucleic acid sequence of about 50-5000 base pairs, the frequency of occurrence of at least one of the selected patterns; and determining whether the occurrence in the test nucleic acid is representative of the occurrence of the selected pattern in the collection of STAR sequences.
  • determining comprises determining whether the frequency of occurrence in the test nucleic acid is representative of the frequency occurrence of the selected pattern in the collection of STAR sequences.
  • the method further comprises determining whether the candidate STAR comprises a gene transcription modulating quality using a method of the invention.
  • the collection of STARS comprises sequence as depicted in SEQ ID NOS:1-119.
  • the invention provides an isolated and/or recombinant nucleic acid sequence comprising a STAR sequence obtainable by a method of the invention.
  • a STAR sequence can exert its activity in a directional way, i.e., more to one side of the fragment containing it than to the other.
  • STAR activity can be amplified in amount by multiplying the number of STAR elements.
  • a STAR element may comprise one or more elements comprising STAR activity.
  • Another way of identifying a sequence capable of conferring STAR activity on a fragment containing it comprises selecting from a vertebrate or plant sequence, a sequence comprising STAR activity and identifying whether the selected sequence and sequences flanking the selected sequence are conserved in another species. Such conserved flanking sequences are likely to be functional sequences.
  • the invention therefore, provides a method for identifying a sequence comprising a STAR element comprising selecting a sequence of about 50 to 5000 base pairs from a vertebrate or plant species comprising a STAR element and identifying whether sequences flanking the selected sequence in the species are conserved in at least one other species.
  • the invention therefore, further provides a method for detecting the presence of a STAR sequence within a nucleic acid sequence of about 50-5000 base pairs, comprising identifying a sequence comprising a STAR sequence in a part of a chromosome of a cell of a species and detecting significant homology between the sequence and a sequence of a chromosome of a different species.
  • the species comprises a plant or vertebrate species, ideally a mammalian species.
  • the invention also provides a method for detecting the presence of a STAR element within a nucleic acid sequence of about 50-5000 base pairs of a vertebrate or plant species, comprising identifying whether a flanking sequence of the nucleic acid sequence is conserved in at least one other species.
  • FIG. 1 is a diagram illustrating the pSelect plasmid used for isolating STAR elements.
  • the zeocin resistance gene is under control of the SV40 promoter, and is upstream of the SV40 polyadenylation site. Upstream of the SV40 promoter is a tandem array of lexA operator sites. Between the lexA operators and the SV40 promoter is a cloning site; test DNAs (e.g., size-fractionated genomic DNA) are cloned into the BamHI site.
  • test DNAs e.g., size-fractionated genomic DNA
  • the plasmid also has the hygromycin resistance gene (hygro) for selection of transformed cells, the EBNA-1 and oriP sequences for plasmid replication in mammalian cells, and the ampicillin resistance gene (ampR) and ori sequence for propagation in Escherichia coli.
  • hygro hygromycin resistance gene
  • ampR ampicillin resistance gene
  • FIG. 2 is a diagram depicting the pSDH-CSP plasmid used for testing STAR activity.
  • the Secreted Alkaline Phosphatase (SEAP) reporter gene is under control of the CMV promoter, and the puromycin resistance selectable marker (puro) is under control of the SV40 promoter. Flanking these two genes are multiple cloning sites (MCSI and MCSII) into which STAR elements can be cloned.
  • the plasmid also has an origin of replication (ori) and ampicillin resistance gene (amp R ) for propagation in Escherichia coli.
  • FIG. 3 is a graph showing that STAR6 (SEQ ID NO:6) and STAR8 (SEQ ID NO:8) improve predictability and yield of transgene expression in U-2 OS cells. Expression of luciferase from the CMV promoter by U-2 OS cells transfected with pSDH-CMV, pSDH-CMV-STAR6, or pSDH-CMV-STAR8 was determined. The STAR-containing constructs confer greater predictability and elevated yield relative to the pSDH-CMV construct alone.
  • FIG. 4 is a graph illustrating that STAR6 (SEQ ID NO:6), but not STAR8 (SEQ ID NO:8), improves predictability and yield of transgene expression in CHO cells.
  • Expression of SEAP from the CMV promoter by CHO cells transfected with pSDH-CSP, pSDH-CSP-STAR6, or pSDH-CSP-STAR8 was determined.
  • the STAR6-containing constructs confer greater predictability and elevated yield relative to the pSDH-CSP construct alone, identifying STAR6 (SEQ ID NO:6) as a promiscuous STAR element.
  • the STAR8-containing constructs do not consistently increase yield or predictability relative to the pSDH-CSP construct, suggesting that STAR8 (SEQ ID NO:8) is a cell line-specific STAR element.
  • FIG. 5 is a graph depicting that STAR6 (SEQ ID NO:6) and STAR49 (SEQ ID NO:49) improve predictability and yield of transgene expression.
  • Expression of SEAP from the CMV promoter by CHO cells transfected with pSDH-CSP, pSDH-CSP-STAR6, or pSDH-CSP-STAR49 was determined.
  • the STAR-containing constructs confer greater predictability and elevated yield relative to the pSDH-CSP construct alone.
  • FIG. 6 is a graph showing the minimal essential sequences of STAR10 (SEQ ID NO:10) and STAR27 (SEQ ID NO:27). Portions of the STAR elements were amplified by PCR: STAR10 (SEQ ID NO:10) was amplified with primers E23 (SEQ ID NO:166) and E12 (SEQ ID NO:155) to yield fragment 10A (corresponding approximately to the first 400 nucleotides of SEQ ID NO:10), E13 (SEQ ID NO:156) and E14 (SEQ ID NO:157) to yield fragment 10B (corresponding approximately to the second 400 nucleotides of SEQ ID NO:10), and E15 (SEQ ID NO:158) and E16 (SEQ ID NO:159) to yield fragment 10C (corresponding approximately to the third 400 nucleotides of SEQ ID NO:10).
  • STAR10 SEQ ID NO:10 was amplified with primers E23 (SEQ ID NO:166) and E12 (SEQ ID NO:155) to yield fragment 10A (corresponding approximately
  • STAR27 was amplified with primers E17 (SEQ ID NO:160) and E18 (SEQ ID NO:161) to yield fragment 27A (corresponding approximately to the first 500 nucleotides of SEQ ID NO:27), E19 (SEQ ID NO:162) and E20 (SEQ ID NO:163) to yield fragment 27B (corresponding to the second 500 nucleotides of SEQ ID NO:27), and E21 (SEQ ID NO:164) and E22 (SEQ ID NO:165) to yield fragment 27C (corresponding approximately to the third 500 nucleotides of SEQ ID NO:27).
  • E17 SEQ ID NO:160
  • E18 SEQ ID NO:161
  • fragment 27A corresponding approximately to the first 500 nucleotides of SEQ ID NO:27
  • E19 SEQ ID NO:162
  • E20 SEQ ID NO:163
  • fragment 27B corresponding to the second 500 nucleotides of SEQ ID NO:27
  • FIG. 7 is a graph illustrating the STAR element function in the context of the SV40 promoter.
  • pSDH-SV40 and pSDH-SV40-STAR6 were transfected into the human osteosarcoma U-2 OS cell line, and expression of luciferase was assayed with or without protection from gene silencing by STAR6 (SEQ ID NO:6) in puromycin-resistant clones.
  • FIG. 8 is a graph showing the STAR element function in the context of the Tet-Off promoter.
  • pSDH-Tet and pSDH-Tet-STAR6 were transfected into the human osteosarcoma U-2 OS cell line, and expression of luciferase was assayed with or without protection from gene silencing by STAR6 (SEQ ID NO:6) in puromycin-resistant clones.
  • FIG. 9 is a schematic diagram of the orientation of STAR elements as they are cloned in the pSelect vector (panel A), as they are cloned into pSDH vectors to preserve their native orientation (panel B), and as they are cloned into pSDH vector in the opposite orientation (panel C).
  • FIG. 10 is a graph showing directionality of STAR66 (SEQ ID NO:66) function.
  • the STAR66 (SEQ ID NO:66) element was cloned into pSDH-Tet in either the native (STAR66 native) or the opposite orientation (STAR66 opposite) and transfected into U-2 OS cells. Luciferase activity was assayed in puromycin resistant clones.
  • FIG. 11 is a southern blot showing copy number-dependence of STAR function. Southern blot of luciferase expression units in pSDH-Tet-STAR10, integrated into U-2 OS genomic DNA. Radioactive luciferase DNA probe was used to detect the amount of transgene DNA in the genome of each clone, which was then quantified with a phosphorimager.
  • FIG. 12 is a graph illustrating copy number-dependence of STAR function.
  • the copy number of pSDH-Tet-STAR10 expression units in each clone was determined by phosphorimagery and compared with the activity of the luciferase reporter enzyme expressed by each clone.
  • FIG. 13 is a schematic diagram and graphs depicting enhancer-blocking and enhancer assays.
  • the luciferase expression vectors used for testing STARs for enhancer-blocking and enhancer activity are shown schematically.
  • the E-box binding site for the E47 enhancer protein is upstream of a cloning site for STAR elements. Downstream of the STAR cloning site is the luciferase gene under control of a human alkaline phosphatase minimal promoter (mp).
  • mp human alkaline phosphatase minimal promoter
  • the histograms indicate the expected outcomes for the three possible experimental situations (see text).
  • Panel A Enhancer-blocking assay.
  • Panel B Enhancer assay.
  • FIG. 14 is a graph showing enhancer-blocking assay. Luciferase expression from a minimal promoter is activated by the E47/E-box enhancer in the empty vector (vector). Insertion of enhancer-blockers (scs, HS4) or STAR elements (STAR elements 1, 2, 3, 6, 10, 11, 18, and 27; SEQ ID NOS:1, 2, 3, 6, 10, 11, 18 and 27, respectively) block luciferase activation by the E47/E-box enhancer.
  • enhancer-blockers scs, HS4
  • STAR elements STAR elements 1, 2, 3, 6, 10, 11, 18, and 27; SEQ ID NOS:1, 2, 3, 6, 10, 11, 18 and 27, respectively
  • FIG. 15 is a graph illustrating enhancer assay. Luciferase expression from a minimal promoter is activated by the E47/E-box enhancer in the empty vector (E47). Insertion of the scs and HS4 elements or various STAR elements (STARs 1, 2, 3, 6, 10, 11, 18, and 27; SEQ ID NOS:1, 2, 3, 6, 10, 11, 18 and 27, respectively) do not activate transcription of the reporter gene.
  • FIG. 16 illustrates STAR18 (SEQ ID NO:18) sequence conservation between mouse and human.
  • the region of the human genome containing 497 base pair STAR18 (SEQ ID NO:18) is shown (black boxes); the element occurs between the HOXD8 and HOXD4 homeobox genes on human chromosome 2. It is aligned with a region in mouse chromosome 2 that shares 72% sequence identity.
  • the region of human chromosome 2 immediately to the left of STAR18 (SEQ ID NO:18) is also highly conserved with mouse chromosome 2 (73% identity; gray boxes); beyond these region, the identity drops below 60%.
  • the ability of these regions from human and mouse, either separately or in combination, to confer growth on zeocin is indicated: ⁇ , no growth; +, moderate growth; ++, vigorous growth; +++, rapid growth.
  • FIG. 17 is a schematic diagram of bio-informatic analysis workflow. For details, see text.
  • FIG. 18 is a schematic diagram showing the results of discriminant analysis on classification of the training set of 65 STAR elements.
  • STAR elements that are correctly classified as STARs by Stepwise Linear Discriminant Analysis (LDA) are shown in a Venn diagram.
  • the variables for LDA were selected from frequency analysis results for hexameric oligonucleotides (“oligos”) and for dyads.
  • the diagram indicates the concordance of the two sets of variables in correctly classifying STARs.
  • FIG. 19 is a graph illustrating that U-2 OS/Tet-Off/lexA-HP1 cells were transfected with candidate Arabidopsis STAR elements and cultivated at low doxycycline concentrations.
  • Total RNA was isolated and subjected to RT-PCR; the bands corresponding to the zeocin and hygromycin resistance mRNAs were detected by Southern blotting and quantified with a phosphorimager. The ratio of the zeocin to hygromycin signals is shown for transfectants containing zeocin expression units flanked by 12 different Arabidopsis STAR elements, the Drosophila scs element, or no flanking element.
  • FIG. 20 is a schematic diagram and graph illustrating that STAR elements improve GFP expression in CHO cells.
  • the ppGIZ and ppGIZ-STAR7 plasmids used for testing STAR activity are shown.
  • the expression unit comprises (from 5′ to 3′) a transgene (encoding for the GFP protein), an IRES, and a selectable marker (zeo, conferring zeocin resistance) under control of the CMV promoter.
  • the expression unit has the SV40 transcriptional terminator at its 3′ end (t).
  • the entire cassette with the expression unit is either flanked by STAR7 (SEQ ID NO:7) elements (STAR7-shielded) or not (Control).
  • the constructs are transfected to CHO-K1 cells.
  • FIG. 21 is a schematic diagram and graph showing that STAR elements improve GFP expression in NSO cells.
  • the ppGIZ and ppGIZ-STAR7 plasmids used for testing STAR activity are shown as in FIG. 20 .
  • the constructs are transfected to NSO cells. Stable colonies are expanded and the GFP signal is determined on a XL-MCL Beckman Coulter flow cytometer. For each independent colony the mean of the GFP signal is plotted. This is taken as measure for the level of GFP expression.
  • the results in FIG. 21 show that in NSO cells the STAR7-shielded (SEQ ID NO:7) construct confers greater predictability and elevated GFP expression relative to the ppGIZ control construct alone.
  • FIG. 22 is a schematic diagram and graph depicting that STAR elements improve GFP expression in 293 cells.
  • the ppGIZ and ppGIZ-STAR7 plasmids used for testing STAR activity are shown as in FIG. 20 .
  • the constructs are transfected to 293 cells. Stable colonies are expanded and the GFP signal is determined on a XL-MCL Beckman Coulter flow cytometer. For each independent colony, the mean of the GFP signal is plotted. This is taken as measure for the level of GFP expression.
  • the results in FIG. 22 show that in 293 cells the STAR7-shielded (SEQ ID NO:7) construct confers greater predictability and elevated GFP expression relative to the ppGIZ control construct alone.
  • quality in relation to a sequence refers to an activity of the sequence.
  • STAR refers to a DNA sequence comprising one or more of the mentioned gene transcription modulating qualities.
  • DNA sequence as used herein, unless otherwise specified, does not refer to a listing of specific ordering of bases, but rather to a physical piece of DNA.
  • a transcription quality with reference to a DNA sequence refers to an effect that the DNA sequence has on transcription of a gene of interest.
  • “Quality” as used herein refers to detectable properties or attributes of a nucleic acid or protein in a transcription system.
  • proteinaceous molecule By a “proteinaceous molecule” is meant herein a molecule comprising amino acids. At least a part of the amino acids are bound to each other to form a peptide. Preferably, the proteinaceous molecule comprises a polypeptide. In this application, the term “proteinaceous molecule” also includes “polypeptide.”
  • essentially the same properties is meant that the properties are essentially the same in kind, not necessarily in amount. For instance, if a proteinaceous molecule has essentially the same properties as a pharmaceutically active compound, the proteinaceous molecule also displays such pharmaceutical activity in kind, not necessarily in amount.
  • a “naturally occurring proteinaceous molecule of the same kind” is meant a proteinaceous molecule with the same primary structure, which is naturally produced in vivo, not influenced by human interference.
  • Examples comprise an antibody produced in vivo by a lymphocyte and erythropoietin produced in vivo by a hepatocyte.
  • host cell and “host cell line” refer to a cell and to homogeneous populations thereof that are capable of expressing a nucleic acid encoding a proteinaceous molecule.
  • recombinant host cell and “recombinant host cell line” refer to a host cell and to homogeneous populations thereof into which a nucleic acid has been introduced.
  • the term “expression” refers to production of a proteinaceous molecule, encoded by a nucleic acid.
  • the production for instance, involves transcription of a DNA sequence, translation of the corresponding mRNA sequence, and/or posttranslational modification.
  • secreted proteins it can also refer to the processes of transcription, translation, and/or post-translational modification (e.g., glycosylation, disulfide bond formation, etc.), followed by exocytosis.
  • multimeric proteins it can include assembly of the multimeric structure from the polypeptide monomers.
  • the term “silencing” refers to diminution of a level of expression of a gene or genes, including transgenes, typically over time.
  • the expression level can be diminished but still detectable, or diminished below the threshold of detection.
  • enhanced expression of a gene encoding a proteinaceous molecule can either involve a higher yield of the proteinaceous molecule, a higher proportion of host cells with acceptable expression levels, and/or a higher stability of a gene expression level.
  • an “affiliated proteinaceous molecule of a cell” is meant a proteinaceous molecule which is naturally produced by the kind of cell in the organism from which the cell is derived.
  • erythropoietin is an affiliated proteinaceous molecule of a hepatocyte, or of a hepatocyte-derived cell line.
  • an antibody is an affiliated proteinaceous molecule of a lymphocyte, or of a lymphocyte-derived cell line, typically of a B-cell or a B-cell derived cell line.
  • a “specific host cell line” is a host cell line derived from a cell which normally expresses a particular proteinaceous molecule (or class of proteinaceous molecules) in the organism from which the cell is derived.
  • heterologous STAR sequence is herein used to define a STAR sequence that is, for example, obtained from a different cell type (from the same species or organism) or is obtained from a different species or organism (either from the same cell type or a different cell type) compared to the cell in which it is used.
  • “Stable” means that the observed transcription level is not significantly changed over at least 30 cell divisions. A stable quality is useful in situations wherein expression characteristics should be predictable over many cell divisions. Typical examples are cell lines transfected with foreign genes. Other examples are transgenic animals and plants and gene therapies. Very often, introduced expression cassettes function differently after increasing numbers of cell divisions or plant or animal generations. A stable quality preferably comprises a capacity to maintain gene transcription in subsequent generations of a transgenic plant or animal. Of course, in the case where expression is inducible, the quality comprises the quality to maintain inducibility of expression in subsequent generations of a transgenic plant or animal.
  • an “acceptable expression level” means an acceptable expression level for commercial exploitation. Whether or not a certain expression level is acceptable for commercial exploitation often depends on the kind of proteinaceous molecule that is produced. Acceptable expression levels of different kinds of proteinaceous molecules often involve different amounts of produced proteinaceous molecule.
  • a STAR sequence, a collection of STAR sequences, and/or a nucleic acid comprising a STAR sequence obtainable by a method of the invention is of course suitable for use in a method of the invention for producing a proteinaceous molecule in a cell.
  • the invention therefore, provides a method for producing a proteinaceous molecule in a cell comprising selecting a cell for its suitability for producing the proteinaceous molecule, providing a nucleic acid encoding the proteinaceous molecule with a nucleic acid comprising a STAR sequence, expressing the resulting nucleic acid in the cell and collecting the proteinaceous molecule, wherein the nucleic acid comprising a STAR sequence is obtainable by a method of the invention for identifying and obtaining a nucleic acid comprising a STAR sequence.
  • the nucleic acid comprising a STAR sequence can be identified and obtained using at least one pattern that is representative for sequences comprising STAR activity.
  • the nucleic acid comprising a STAR sequence is identified and obtained using at least one pattern as depicted in Table 6 (SEQ ID NOS:177-342) or Table 7 (SEQ ID NOS:343-1072).
  • a cell can be selected for its suitability for producing a proteinaceous molecule in many different ways. For instance, it can be determined whether the cell is competent of nucleic acid uptake. A nucleic acid encoding a proteinaceous molecule is preferably easily introduced into the cell. Furthermore it can be determined whether the cell secretes produced proteinaceous molecule. Secreted proteinaceous molecule can usually be easily collected. Collecting proteinaceous molecules that are not secreted usually involves sacrificing at least part of a culture. This implicates separating a proteinaceous molecule of interest from other cell components, optionally starting up a new culture, etc. This is more cumbersome. Hence, in a preferred embodiment, a method of the invention is provided wherein the proteinaceous molecule is secreted by the cell.
  • a method of the invention wherein the cell is selected for its capability of post-translationally modifying the proteinaceous molecule, such that the proteinaceous molecule has essentially the same properties as a naturally occurring proteinaceous molecule of the same kind.
  • the same proteinaceous molecule can be modified differently when produced in different host cells. These differences can affect the properties of such proteinaceous molecules, such as for instance pharmaceutical properties. It is, therefore, highly preferred to select a cell which produces a proteinaceous molecule with essentially the same properties as its naturally occurring counterpart.
  • a proteinaceous molecule can be produced that is physically different from its natural counterpart, but which is functionally essentially the same.
  • a cell is selected for its capability of post-translationally modifying the proteinaceous molecule in essentially the same way as the proteinaceous molecule is post-translationally modified in nature.
  • the invention provides a method of the invention wherein the proteinaceous molecule comprises an affiliated proteinaceous molecule of the cell.
  • a host cell or a cell line derived from a cell, which normally produces a proteinaceous molecule of interest in the organism from which the cell is derived.
  • These cells are capable of carrying out post-translational modifications of their affiliated proteinaceous molecules such that the resulting proteinaceous molecule has the same kind of properties in kind, not necessarily in amount, as a proteinaceous molecule of the same kind which is normally present in the organism.
  • Such specific cells are naturally adapted for production of the affiliated proteinaceous molecule.
  • Apart from other activities of STAR sequences it is also possible to at least partly solve a problem of low production of proteinaceous molecules, which often occurs with such specific cells.
  • Providing at least one STAR sequence to a nucleic acid encoding such proteinaceous molecule will enhance production (yield) of the proteinaceous molecule by the specific cell, increase the proportion of host cells with acceptable expression levels, and/or increase stability of a gene expression level.
  • a method of the invention wherein the cell is selected for suitable growth in a suspension culture. This facilitates culturing of the cell, and collection of produced proteinaceous molecule, especially when the proteinaceous molecule is secreted into the suspension. More preferably, the cell is selected for suitable growth in a serum-free culture, since serum can contain contaminants and pathogens. Such contaminants/pathogens often have to be separated from produced proteinaceous molecule. This requires an extra step, which consumes time and chemicals, with a potential loss of produced proteinaceous molecule. Moreover, a possibility of a presence of pathogens involves a potential risk for employees. If a pathogen has been present in a culture, a produced proteinaceous molecule is not allowed to be used anymore for commercial and/or medical applications.
  • the cell is selected for the presence of an adenovirus Early Region 1 (E1) sequence.
  • E1 adenovirus Early Region 1
  • the presence of an adenoviral E1 sequence enhances protein production in a cell.
  • an adenoviral E1 sequence is suitable for host cells engineered for protein production.
  • the invention also provides a method for producing a proteinaceous molecule comprising:
  • the method is performed with a host cell whose genome has not been modified by human interference.
  • the method then results in enhanced expression of a proteinaceous molecule which is encoded by the genome of the host cell.
  • the proteinaceous molecule may be normally expressed by the host cell in the organism from which the cell is derived, but it may also normally be subject to silencing, resulting in little or no expression in the host cell under normal conditions.
  • Introduction of at least one STAR sequence can at least in part inhibit silencing of a gene of interest induced by gene-transcription repressing chromatin. Expression of a proteinaceous molecule is enhanced by introduction of a STAR sequence.
  • the host cell is transfected with a nucleic acid of interest.
  • nucleic acid for instance, encodes a heterologous proteinaceous molecule which is not naturally encoded by the genome of the host cell.
  • Introduction of a STAR sequence also enhances expression of such heterologous proteinaceous molecule.
  • the STAR sequence can be introduced randomly into the genome of the host cell, using methods known in the art (for instance calcium precipitation, transfection with a vector comprising a nucleic acid of interest, use of a gene delivery vehicle, etc). If a STAR sequence is introduced near a nucleic acid sequence encoding a proteinaceous molecule, it is capable of enhancing expression of the proteinaceous molecule. Cells expressing a desired proteinaceous molecule can be isolated from cultures with randomly inserted STAR sequences.
  • the STAR sequence is introduced into the host cell by homologous recombination.
  • a nucleic acid comprising a STAR sequence can be provided with an additional sequence.
  • the additional sequence can be chosen such that it is at least in part homologous to a nucleic acid sequence in the host cell which is known to be present in vicinity of a gene encoding a proteinaceous molecule of interest. If a nucleic acid comprising a STAR sequence and such additional sequence is provided to the host cell, it can be incorporated into the host cell's genome by homologous recombination at the site with the (partly) homologous nucleic acid sequence. As a result, the STAR sequence is introduced in vicinity of the gene encoding the proteinaceous molecule of interest. Expression of the proteinaceous molecule is then enhanced by the introduced STAR sequence.
  • a preferred embodiment of the present invention provides a method of the invention wherein the STAR sequence comprises a species-specific STAR sequence. More preferably, the STAR sequence comprises a cell type-specific STAR sequence.
  • STAR elements Two types have been identified. Promiscuous STAR elements are able to function in more than one host cell line. For example, STAR6 (SEQ ID NO:6) increases the predictability, yield, and stability of a transgene in both the U-2 OS human osteosarcoma cell line and in CHO (Chinese hamster ovary) cells. Other STAR elements are species-specific and/or cell type-specific; for example, STAR8 (SEQ ID NO:8) increases the predictability, yield, and stability of transgenes in U-2 OS cells, but not in CHO cells (see Examples 2 and 3 and FIGS. 3 and 4 ).
  • STAR sequence which is naturally present in the cell can be used in a method of the invention.
  • STAR sequence is referred to as a cell type-specific STAR sequence.
  • a STAR sequence which is naturally present in a species from which the cell is derived can also be used.
  • Such STAR sequence is referred to as a species-specific STAR sequence.
  • a species-specific STAR sequence may be naturally present in the cell type, although this is not necessary.
  • a known species-specific STAR sequence or cell-type specific STAR sequence can be used in a method of the invention.
  • a (previously unknown) species-specific STAR sequence or cell-type specific STAR sequence can be detected and isolated by a method as described by the present inventors (EP 01202581.3).
  • the use of a species-specific STAR sequence or cell type-specific STAR sequence is preferred because such sequence is especially active in the host cell and is adapted to the specific circumstances within the cell.
  • such cell type-specific STAR sequence may interact with a protein which is not present in some other cell-types. In that case, the cell type-specific STAR sequence will be less capable—if at all—of enhancing expression in cells lacking the protein.
  • a species-specific or cell type-specific STAR element often has functional characteristics that are superior to promiscuous STAR elements.
  • a cell line-specific STAR element can satisfy product safety or ethical considerations for use of the host cell line.
  • a promiscuous STAR sequence is particularly useful if no tissue specific or cell-type specific STAR sequence is known. In that case a known promiscuous STAR sequence can be used. This saves efforts to detect and isolate a cell-type specific STAR sequence.
  • STAR sequences are listed in SEQ ID NOS:1-119.
  • a method of the invention is provided wherein the STAR sequence comprises a sequence as depicted in SEQ ID NOS:1-119.
  • the invention provides a cell line that comprises at least one heterologous STAR sequence or a functional equivalent and/or a functional fragment thereof.
  • the cell line is a human cell line.
  • the invention provides multiple examples of STAR sequences and also methods of testing STAR sequences and hence, a person skilled in the art is very well capable of obtaining a functional equivalent and/or a functional fragment of a STAR sequence, for example by deletion or mutation.
  • the invention provides a non-human cell line that comprises at least one recombinant STAR sequence derived or obtained from a human cell, i.e., a human STAR sequence. It is clear that the amount of STAR sequences may vary, for example, a cell line according to the invention may comprise two, three, or four, or even more STAR sequences which may either be identical or different from each other.
  • the invention provides a cell line provided with a nucleic acid comprising a STAR sequence, wherein the cell line is selected for its suitability for producing a proteinaceous molecule.
  • a cell line of the invention comprises a vertebrate or plant cell line.
  • a vertebrate cell line is very suitable for producing a human proteinaceous molecule of interest, because vertebrates are phylogenetically close related.
  • Plant cells are for instance very suitable for vaccine production.
  • Vaccine production in plants can be inexpensive, while the vaccine can be easily delivered to an individual by eating the edible portion of the plant (Mercenier et al., 2001).
  • a cell line of the invention is particularly suitable for production of a proteinaceous molecule of interest, because the STAR sequence can enhance expression of a gene of interest (higher yield of a proteinaceous molecule, higher proportion of host cells with acceptable expression levels, and/or higher stability of a gene expression level).
  • Methods for generating a cell line are known in the art and many techniques are known to provide a cell with a nucleic acid of interest.
  • many general purpose cell lines are available. Such cell lines can be dedicated to production of a certain proteinaceous molecule using recombinant techniques. Examples of available cell lines include CHO cells from Chinese hamster ovary and BHK cells from baby hamster kidney (as described above).
  • Another embodiment of the invention provides a cell line provided with a nucleic acid comprising a STAR sequence, wherein the cell line comprises an adenovirus Early Region 1 sequence.
  • an adenoviral E1 sequence enhances cellular protein production.
  • a cell line of the invention is provided wherein the cell line comprises a U-2 OS osteosarcoma, CHO, 293, HuNS-1 myeloma, WERI-Rb-1 retinoblastoma, BHK, Vero, non-secreting mouse myeloma Sp2/0-Ag 14, non-secreting mouse myeloma NSO, or NCI-H295R adrenal gland carcinoma (ATCC CRL-2128) cell line.
  • a cell line of the invention is particularly suitable for production of a proteinaceous molecule, because production can be enhanced by one or more STAR sequences (higher yield of a proteinaceous molecule, higher proportion of host cells with acceptable expression levels, and/or higher stability of a gene expression level).
  • a cell line of the invention can comprise promiscuous, species-specific and/or cell type-specific STAR sequences.
  • a cell line of the invention can be used to produce a heterologous proteinaceous molecule, and/or an affiliated proteinaceous molecule.
  • a use of a cell line of the invention for the production of a proteinaceous molecule is also herewith provided.
  • the proteinaceous molecule comprises an affiliated protein of the cell line.
  • the invention provides a method for selecting a cell suitable for producing a proteinaceous molecule comprising:
  • the desired property for instance, comprises a pharmaceutical property.
  • the property can be influenced by post translational modification(s), a configuration of a produced proteinaceous molecule, etc.
  • the invention provides a method for selecting a cell suitable for producing a proteinaceous molecule comprising:
  • the nucleic acid comprising a STAR sequence can be randomly introduced into the genome of the host cell.
  • the nucleic acid sequence is introduced into the genome of the host cell by homologous recombination.
  • STAR elements are identified and cloned from human genomic DNA based on their ability to block the spread of transcriptional repression from DNA binding sites for repressor proteins in a test vector, as described in this example.
  • the method described in this example is applicable in principle to any mammalian cell line, for isolation of both promiscuous and cell line-specific STAR elements.
  • the selection vector for STAR elements, pSelect-SV40-zeo (“pSelect,” FIG. 1 ) was constructed as follows: the pREP4 vector (Invitrogen V004-50) was used as the plasmid backbone. It provides the Epstein Barr oriP origin of replication and EBNA-1 nuclear antigen for high-copy episomal replication in primate cell lines; the hygromycin resistance gene with the thymidine kinase promoter and polyadenylation site, for selection in mammalian cells; and the ampicillin resistance gene and colE1 origin of replication for maintenance in Escherichia coli .
  • the vector contains four consecutive LexA operator sites between XbaI and NheI restriction sites (Bunker and Singer, 1994). Embedded between the LexA operators and the NheI site is a polylinker consisting of the following restriction sites: HindIII-AscI-BamHI-AscI-HindIII. Between the NheI site and a SalI site is the zeocin resistance gene with the SV40 promoter and polyadenylation site, derived from pSV40/Zeo (Invitrogen V502-20); this is the selectable marker for the STAR screen.
  • Gene libraries were constructed by Sau3AI digestion of human genomic DNA, either purified from placenta (Clontech 6550-1) or carried in bacterial/P1 (BAC/PAC) artificial chromosomes.
  • the BAC/PAC clones contain genomic DNA from the 1q12 cytogenetic region (clones RP1154H19 and RP3328E19), from the HOX cluster of homeotic genes (clones RP1167F23, RP1170019, and RP11387A1), or from human chromosome 22 (Research Genetics 96010-22).
  • the DNAs were size-fractionated, and the 0.5-2 kb size fraction ligated into BamHI-digested pSelect vector, by standard techniques (Sambrook et al., 1989).
  • U-2 OS human osteosarcoma cell line
  • U-2 OS was stably transfected with the pTet-Off plasmid (Clontech K1620-A), encoding a protein chimera consisting of the Tet-repressor DNA binding domain and the VP16 transactivation domain.
  • the cell line was subsequently stably transfected with fusion protein genes containing the LexA DNA binding domain, and the coding regions of HP1, MeCP2, or HPC2 (three Drosophila proteins that repress gene expression when tethered to DNA).
  • the LexA-repressor genes are under control of the Tet-Off transcriptional regulatory system (Gossen and Bujard, 1992).
  • Zeocin-resistant colonies from the library transfection were propagated, and plasmid DNA isolated and rescued into E. coli by standard techniques (Sambrook et al., 1989).
  • the candidate STAR elements in the rescued DNA were analyzed by restriction endonuclease mapping (Sambrook et al., 1989), and tested for STAR activity (zeocin resistance) after re-transfection to U-2 OS/Tet-Off/LexA-repressor cells and lowering the doxycycline concentration.
  • the human genomic DNA inserts in these plasmids were sequenced by the dideoxy method (Sanger et al., 1977) using a Beckman CEQTM2000 automated DNA sequencer, using the manufacturer's instructions. Briefly, DNA was purified from E. coli using QIAprep® Spin Miniprep and Plasmid Midi Kits (QIAGEN® 27106 and 12145, respectively). Cycle sequencing was carried out using custom oligonucleotides corresponding to the pSelect vector (primers D89 (SEQ ID NO:149) and D95 (SEQ ID NO:154); all oligonucleotides are described in Table 2), in the presence of dye terminators (CEQTM Dye Terminator Cycle Sequencing Kit, Beckman 608000).
  • STAR DNA sequences were located in the human genome using BLAST (Basic Local Alignment Search Tool (Altschul et al., 1990); worldwideweb.ncbi.nlm.nih.gov/BLAST/).
  • the screens of human genomic DNA have yielded 66 STAR elements; the lengths and chromosomal locations of these elements are tabulated in SEQ ID NO:1-SEQ ID NO:66. They confer zeocin resistance on U-2 OS host cells when placed between LexA-repressor binding sites and the zeocin resistance gene. Their anti-repression activity was demonstrated both in the initial screen and upon re-transfection (demonstrating that the anti-repression activity is due to the STAR element and not to somatic acquisition of zeocin resistance).
  • the STAR elements correspond to known and unique sequences in the human genome, as demonstrated by BLAST searches (Table 3). In some cases, the cloned element is a chimera of two unlinked genomic loci (e.g., STAR3 (SEQ ID NO:3), Table 3). They range in length from 500 to 2361 base pairs in length.
  • STAR elements function to block the effect of transcriptional repression influences on transgene expression units. These repression influences can be due to heterochromatin (“position effects”) or to adjacent copies of the transgene (“repeat-induced gene silencing”).
  • position effects can be due to heterochromatin
  • repeat-induced gene silencing can be due to adjacent copies of the transgene.
  • Two of the benefits of STAR elements for heterologous protein production are increased predictability of finding high-expressing primary recombinant host cells and increased yield during production cycles. These benefits are illustrated in this example.
  • the pSDH-Tet vector was constructed by polymerase chain reaction amplification (PCR) of the luciferase open reading frame from plasmid pREP4-HSF-Luc (van der Vlag et al., 2000) using primers C67 (SEQ ID NO:136) and C68 (SEQ ID NO:137), and insertion of the SacII/BamHI fragment into SacII/BamHI-digested pUHD10-3 (Gossen and Bujard, 1992).
  • PCR polymerase chain reaction amplification
  • the luciferase expression unit was re-amplified with primers C65 (SEQ ID NO:134) and C66 (SEQ ID NO:135), and re-inserted into pUHD10-3 in order to flank it with multiple cloning sites (MCSI and MCSII).
  • An AscI site was then introduced into MCSI by digestion with EcoRI and insertion of a linker (comprised of annealed oligonucleotides D93 (SEQ ID NO:152) and D94 (SEQ ID NO:153)).
  • the CMV promoter was amplified from plasmid pCMV-Bsd (Invitrogen K510-01) with primers D90 (SEQ ID NO:150) and D91 (SEQ ID NO:151), and used to replace the Tet-Off promoter in pSDH-Tet by SalI/SacII digestion and ligation to create vector pSDH-CMV.
  • the luciferase open reading frame in this vector was replaced by SEAP (Secreted Alkaline Phosphatase) as follows: vector pSDH-CMV was digested with SacII and BamHI and made blunt; the SEAP open reading frame was isolated from pSEAP-basic (Clontech 6037-1) by EcoRI/SalI digestion, made blunt and ligated into pSDH-CMV to create vector pSDH-CS.
  • the puromycin resistance gene under control of the SV40 promoter was isolated from plasmid pBabe-Puro (Morgenstern and Land, 1990) by PCR, using primers C81 (SEQ ID NO:138) and C82 (SEQ ID NO:139).
  • STAR elements were inserted into MCSI and MCSII in two steps, by digestion of the STAR element and the pSDH-CSP vector with an appropriate restriction enzyme, followed by ligation.
  • the orientation of STAR elements in recombinant pSDH vectors was determined by restriction mapping, and in all cases verified by DNA sequence analysis using primers C85 (SEQ ID NO:140), E42 (SEQ ID NO:168), and E25 (SEQ ID NO:167) (Table 2; see Example 1).
  • Luciferase assay Luciferase activity (Himes and Shannon, 2000) was assayed in resuspended cells according to the instructions of the assay kit manufacturer (Roche 1669893), using a luminometer (Turner 20/20TD). Total cellular protein concentration was determined by the bicinchoninic acid method according to the manufacturer's instructions (Sigma B-9643), and used to normalize the luciferase data.
  • CHO-K1 Chinese Hamster Ovary cell line CHO-K1 (ATCC CCL-61) was cultured in HAMS-F12 medium+10% Fetal Calf Serum containing 2 mM glutamine, 100 U/ml penicillin, and 100 micrograms/ml streptomycin at 37° C./5% CO 2 .
  • Cells were transfected with recombinant pSDH-CSP vectors using SuperFect® (QIAGEN®) as described by the manufacturer. Briefly, cells were seeded to culture vessels and grown overnight to 70-90% confluence.
  • SuperFect® reagent was combined with plasmid DNA (linearized in this example by digestion with PvuI) at a ratio of 6 microliters per microgram (e.g., for a 10 cm Petri dish, 20 micrograms DNA and 120 microliters SuperFect®) and added to the cells. After overnight incubation, the transfection mixture was replaced with fresh medium, and the transfected cells were incubated further. After overnight cultivation, 5 micrograms/ml puromycin was added. Puromycin selection was complete in two weeks, after which time individual puromycin resistant CHO/pSDH-CSP clones were isolated at random and cultured further.
  • SEAP activity (Berger et al., 1988, Henthorn et al., 1988, Kain, 1997, Yang et al., 1997) in the culture medium of CHO/pSDH-CSP clones was determined as described by the manufacturer (Clontech Great EscAPe kit #K2041). Briefly, an aliquot of medium was heat inactivated at 65° C., then combined with assay buffer and CSPD chemiluminescent substrate and incubated at room temperature for ten minutes. The rate of substrate conversion was then determined in a luminometer (Turner 20/20TD). Cell density was determined by counting trypsinized cells in a Coulter ACT10 cell counter. Luminescence units were converted into picograms SEAP based on a SEAP positive control calibration curve, and normalized to cell number.
  • the recombinant U-2 OS clones with STAR6 (SEQ ID NO:6) flanking the expression units had higher yields than the STAR-less clones: the STAR6 clones had maximal luciferase expression levels five-fold higher than the STAR-less clones.
  • clones with the STAR6 (SEQ ID NO:6) element in the expression units were isolated that express two- to three-fold higher SEAP activity than clones whose expression units do not include this STAR element.
  • STAR6-containing (SEQ ID NO:6-containing) clones that express SEAP activity at or above the maximal activity of the STAR-less clones is quite high: 40% of the STAR6 clone populations exceed the highest SEAP expression of the pSDH-CSP clones.
  • STAR6 SEQ ID NO:6
  • STAR6 SEQ ID NO:6
  • STAR6 SEQ ID NO:6
  • the cell lines used are derived from different species (human and hamster) and different tissue types (bone and ovary), reflecting the broad range of host cells in which this STAR element can be utilized in improving heterologous protein expression.
  • STAR8 (SEQ ID NO:8) is a Cell Line-Specific STAR Element
  • the patterns of gene expression and epigenetic gene regulation in a host cell line reflect the developmental state of the somatic cells from which they are derived. Furthermore, the biotechnology industry takes advantage of general purpose cell lines from different species according to specific requirements of a heterologous protein production process. Therefore, it is expected that some STAR elements will not function in cell lines other than those in which they are isolated. This expectation has been fulfilled by some of the STAR elements shown in Table 3. One example will be given here.
  • the recombinant U-2 OS clones with the STAR8 (SEQ ID NO:8) element flanking the expression units had higher yields than the STAR-less clones: the highest expression observed from STAR8 clones was two- to three-fold higher than the expression from STAR-less clones.
  • the STAR8 (SEQ ID NO:8) element conferred greater predictability as well: for this STAR element, ⁇ 15% of the clones displayed luciferase expression at levels comparable to or greater than the STAR-less clone with the highest expression level.
  • one clone with the STAR8 (SEQ ID NO:8) element in the expression unit had a yield approximately two-fold higher than the highest-expressing STAR-less clone.
  • STAR8 SEQ ID NO:8
  • U-2 OS cell line in which STAR8 (SEQ ID NO:8) was originally isolated is not an accurate predictor of its performance in other cell lines.
  • STAR8 SEQ ID NO:8 is thus an example of a cell line-specific STAR element.
  • the U-2 OS cell line was transfected with the plasmid pSDH-Tet-STAR6 and cultivated as described in Example 2. Individual puromycin-resistant clones were isolated and cultivated further in the absence of doxycycline. At weekly intervals the cells were transferred to fresh culture vessels at a dilution of 1:20. Luciferase activity was measured at periodic intervals as described in Example 2. After 15 weeks, the cultures were divided into two replicates; one replicate continued to receive puromycin, while the other replicate received no antibiotic for the remainder of the experiment (25 weeks total).
  • Table 4 presents the data on luciferase expression by an expression unit flanked with STAR6 (SEQ ID NO:6) during prolonged growth with or without antibiotic.
  • STAR6 SEQ ID NO:6
  • the expression of the reporter transgene, luciferase remains stable in the U-2 OS host cells for the duration of the experiment.
  • the expression of luciferase was essentially stable in the absence of antibiotic selection.
  • This demonstrates the ability of STAR elements to protect transgenes from silencing or loss during prolonged cultivation. It also demonstrates that this property is independent of antibiotic selection. Therefore, production of heterologous proteins is possible without incurring the costs of the antibiotic or of difficult downstream processing.
  • STAR elements are isolated from the genetic screen described in Example 1.
  • the screen uses libraries constructed with human genomic DNA that was size-fractionated to approximately 0.5-2 kilobases (supra).
  • the STAR elements range from 500 to 2361 base pairs (Table 3). It is likely that, for many of the STAR elements that have been isolated, STAR activity is conferred by a smaller DNA fragment than the initially isolated clone. It is useful to determine these minimum fragment sizes that are essential for STAR activity, for two reasons. First, smaller functional STAR elements would be advantageous in the design of compact expression vectors, since smaller vectors transfect host cells with higher efficiency. Second, determining minimum essential STAR sequences permits the modification of those sequences for enhanced functionality. Two STAR elements have been fine-mapped to determine their minimal essential sequences.
  • STAR10 SEQ ID NO:10) (1167 base pairs
  • STAR27 SEQ ID NO:27
  • STAR10 SEQ ID NO:10
  • STAR27 SEQ ID NO:27
  • STAR10 SEQ ID NO:10
  • 10A ⁇ 400 base pairs, corresponding to approximately the first 400 nucleotides of SEQ ID NO:10 confers on transfected cells vigorous growth in the presence of zeocin, exceeding that of the full-length STAR element.
  • Cells transfected with pSelect constructs containing the other two sub-fragments do not grow in the presence of zeocin.
  • STAR27 confers moderate growth in zeocin to transfected cells in this experiment ( FIG. 6 ).
  • One of the sub-fragments of this STAR, 27B ( ⁇ 500 base pairs, corresponding to approximately the second 500 nucleotides of SEQ ID NO:27), permits weak growth of the host cells in zeocin-containing medium. This suggests that the anti-repression activity of this STAR is partially localized on sub-fragment 27B, but full activity requires sequences from 27A (corresponding to approximately the first 500 nucleotides of SEQ ID NO:27) and/or 27C (corresponding to approximately the third 500 nucleotides of SEQ ID NO:27) (each ⁇ 500 base pairs) as well.
  • Transgene transcription is achieved by placing the transgene open reading frame under control of an exogenous promoter.
  • the choice of promoter is influenced by the nature of the heterologous protein and the production system. In most cases, strong constitutive promoters are preferred because of the high yields they can provide. Some viral promoters have these properties; the promoter/enhancer of the cytomegalovirus immediate early gene (“CMV promoter”) is generally regarded as the strongest promoter in common biotechnological use (Boshart et al., 1985, Doll et al., 1996, Foecking and Hofstetter, 1986).
  • CMV promoter cytomegalovirus immediate early gene
  • the simian virus SV40 promoter is also moderately strong (Boshart et al., 1985, Foecking and Hofstetter, 1986) and is frequently used for ectopic expression in mammalian cell vectors.
  • the Tet-Off promoter is inducible: the promoter is repressed in the presence of tetracycline or related antibiotics (doxycycline is commonly used) in cell-lines which express the tTA plasmid (Clontech K1620-A), and removal of the antibiotic results in transcriptional induction (Deuschle et al., 1995, Gossen and Bujard, 1992, Izumi and Gilbert, 1999, Umana et al., 1999).
  • pSDH-SV40 was constructed by PCR amplification of the SV40 promoter (primers D41 (SEQ ID NO:142) and D42 (SEQ ID NO:143)) from plasmid pSelect-SV40-Zeo (Example 1), followed by digestion of the PCR product with SacII and SalI.
  • the pSDH-CMV vector was digested with SacII and SalI to remove the CMV promoter, and the vector and SV40 fragment were ligated together to create pSDH-SV40.
  • STAR6 SEQ ID NO:6 was cloned into MCSI and MCSII as described in Example 2.
  • the plasmids pSDH-Tet, pSDH-Tet-STAR6, pSDH-Tet-STAR7, pSDH-SV40 and pSDH-SV40-STAR6 were co-transfected with pBabe-Puro into U-2 OS using SuperFect® as described by the manufacturer. Cell cultivation, puromycin selection, and luciferase assays were carried out as described in Example 2.
  • FIGS. 3 , 7 , and 8 compare the expression of the luciferase reporter gene from three different promoters: two strong and constitutive viral promoters (CMV and SV40), and the inducible Tet-Off promoter. All three promoters were tested in the context of the STAR6 (SEQ ID NO:6) element in U-2 OS cells. The results demonstrate that the yield and predictability from all three promoters are increased by STAR6 (SEQ ID NO:6). As described in Examples 2 and 5, STAR6 (SEQ ID NO:6) is beneficial in the context of the CMV promoter ( FIG. 3 ). Similar improvements are seen in the context of the SV40 promoter ( FIG.
  • STAR Element Function can be Directional
  • nucleic acid sequences can be symmetrical (e.g., palindromic), longer, naturally-occurring sequences are typically asymmetrical.
  • information content of nucleic acid sequences is directional and the sequences themselves can be described with respect to their 5′ and 3′ ends.
  • the directionality of nucleic acid sequence information affects the arrangement in which recombinant DNA molecules are assembled using standard cloning techniques known in the art (Sambrook et al., 1989).
  • STAR elements are long, asymmetrical DNA sequences, and have a directionality based on the orientation in which they were originally cloned in the pSelect vector.
  • the STAR66 (SEQ ID NO:66) element was cloned into pSDH-Tet as described in Example 2.
  • U-2 OS cells were co-transfected with plasmids pSDH-Tet-STAR66-native and pSDH-Tet-STAR66-opposite, and cultivated as described in Example 2. Individual clones were isolated and cultivated; the level of luciferase expression was determined as described (supra).
  • STAR66 SEQ ID NO:66
  • the yield of the highest-expressing clone when STAR66 (SEQ ID NO:66) is in the native orientation is considerably higher (100 luciferase units) and the predictability is much higher, as well: seven clones of the native-orientation population (30%) express luciferase above the level of the highest-expressing clone from the opposite-orientation population, and 15 of the clones in the native-orientation population (60%) express luciferase above ten relative luciferase units. Therefore, it is demonstrated that STAR66 (SEQ ID NO:66) function is directional.
  • Transgene expression units for heterologous protein expression are generally integrated into the genome of the host cell to ensure stable retention during cell division. Integration can result in one or multiple copies of the expression unit being inserted into the genome; multiple copies may or may not be present as tandem arrays.
  • the increased yield demonstrated for transgenes protected by STAR elements suggests that STAR elements are able to permit the transgene expression units to function independently of influences on transcription associated with the site of integration in the genome (independence from position effects (Boivin and Dura, 1998)). It suggests further that the STAR elements permit each expression unit to function independently of neighboring copies of the expression unit when they are integrated as a tandem array (independence from repeat-induced gene silencing (Garrick et al., 1998)). Copy number-dependence is determined from the relationship between transgene expression levels and copy number, as described in the example below.
  • U-2 OS cells were co-transfected with pSDH-Tet-STAR10 and cultivated under puromycin selection as described (supra). Eight individual clones were isolated and cultivated further. Then cells were harvested, and one portion was assayed for luciferase activity as described (supra). The remaining cells were lysed and the genomic DNA purified using the DNeasy® Tissue Kit (QIAGEN® 69504) as described by the manufacturer. DNA samples were quantitated by UV spectrophotometry. Three micrograms of each genomic DNA sample were digested with PvuII and XhoI overnight as described by the manufacturer (New England Biolabs), and resolved by agarose gel electrophoresis.
  • DNA fragments were transferred to a nylon membrane as described (Sambrook et al., 1989), and hybridized with a radioactively labeled probe to the luciferase gene (isolated from BamHI/SacII-digested pSDH-Tet). The blot was washed as described (Sambrook et al., 1989) and exposed to a phosphorimager screen (Personal F/X, BioRad). The resulting autoradiogram ( FIG. 11 ) was analyzed by densitometry to determine the relative strength of the luciferase DNA bands, which represents the transgene copy number.
  • the enzyme activities and copy numbers (DNA band intensities) of luciferase in the clones from the pSDH-Tet-STAR10 clone population is shown in FIG. 12 .
  • Enhancers are characteristically able to affect promoters even when they are located far away (many kilobase pairs) from the promoter.
  • Negative influences that act by heterochromatin formation e.g., Polycomb group proteins
  • the biochemical basis for enhancer function and for heterochromatin formation is fundamentally similar, since they both involve binding of proteins to DNA. Therefore, it is important to determine whether STAR elements are able to block positive influences as well as negative influences, in other words, to shield transgenes from genomic enhancers in the vicinity of the site of integration.
  • the ability to shield transgenes from enhancer activity ensures stable and predictable performance of transgenes in biotechnological applications. This example examines the performance of STAR elements in an enhancer-blocking assay.
  • STAR activity Another feature of STAR activity that is important to their function is the increased yield they confer on transgenes (Example 2).
  • STARs are isolated on the basis of their ability to maintain high levels of zeocin expression when heterochromatin-forming proteins are bound adjacent to the candidate STAR elements. High expression is predicted to occur because STARs are anticipated to block the spread of heterochromatin into the zeocin expression unit.
  • a second scenario is that the DNA fragments in zeocin-resistant clones contain enhancers. Enhancers have been demonstrated to have the ability to overcome the repressive effects of Polycomb-group proteins such as those used in the method of the STAR screen (Zink and Paro, 1995). Enhancers isolated by this phenomenon would be considered false positives, since enhancers do not have the properties claimed here for STARs. In order to demonstrate that STAR elements are not enhancers, they have been tested in an enhancer assay.
  • the enhancer-blocking assay and the enhancer assay are methodologically and conceptually similar.
  • the assays are shown schematically in FIG. 13 .
  • the ability of STAR elements to block enhancers is performed using the E47/E-box enhancer system.
  • the E47 protein is able to activate transcription by promoters when it is bound to an E-box DNA sequence located in the vicinity of those promoters (Quong et al., 2002).
  • E47 is normally involved in regulation of B and T lymphocyte differentiation (Quong et al., 2002), but it is able to function in diverse cell types when expressed ectopically (Petersson et al., 2002).
  • the E-box is a palindromic DNA sequence, CANNTG (Knofler et al., 2002).
  • an E-box is placed upstream of a luciferase reporter gene (including a minimal promoter) in an expression vector.
  • a cloning site for STAR elements is placed between the E-box and the promoter.
  • the E47 protein is encoded on a second plasmid. The assay is performed by transfecting both the E47 plasmid and the luciferase expression vector into cells; the E47 protein is expressed and binds to the E-box, and the E47/E-box complex is able to act as an enhancer.
  • the E47/E-box complex enhances luciferase expression ( FIG. 13A , situation 1).
  • STAR elements are inserted between the E-box and the promoter, their ability to block the enhancer is demonstrated by reduced expression of luciferase activity ( FIG. 13A , situation 2); if STARs cannot block enhancers, luciferase expression is activated ( FIG. 13A , situation 3).
  • STAR elements to act as enhancers utilizes the same luciferase expression vector. In the absence of E47, the E-box itself does not affect transcription. Instead, enhancer behavior by STAR elements will result in activation of luciferase transcription.
  • the assay is performed by transfecting the luciferase expression vector without the E47 plasmid. When the expression vector does not contain STAR elements, luciferase expression is low ( FIG. 13B , situation 1). If STAR elements do not have enhancer properties, luciferase expression is low when a STAR element is present in the vector ( FIG. 13B , situation 2). If STAR elements do have enhancer properties, luciferase expression will be activated in the STAR-containing vectors ( FIG. 13B , situation 3).
  • the luciferase expression vector was constructed by inserting the E-box and a human alkaline phosphatase minimal promoter from plasmid mu-E5+E2 ⁇ 6-cat(x) (Ruezinsky et al., 1991) upstream of the luciferase gene in plasmid pGL3-basic (Promega E1751), to create pGL3-E-box-luciferase (gift of W. Romanow).
  • the E47 expression plasmid contains the E47 open reading frame under control of a beta-actin promoter in the pHBAPr-1-neo plasmid; E47 in constitutively expressed from this plasmid (gift of W. Romanow).
  • STAR elements 1, 2, 3, 6, 10, 11, 18, and 27 have been cloned into the luciferase expression vector.
  • Clones containing the Drosophila scs element and the chicken beta-globin HS4-6 ⁇ core (“HS4”) element have been included as positive controls (they are known to block enhancers, and to have no intrinsic enhancer properties (Chung et al., 1993, Kellum and Schedl, 1992)), and the empty luciferase expression vector has been included as a negative control. All assays were performed using the U-2 OS cell line.
  • the E47 plasmid was co-transfected with the luciferase expression vectors (empty vector, or containing STAR or positive-control elements).
  • the E47 plasmid was co-transfected with STARless luciferase expression vector as a positive control for enhancer activity; all other samples received a mock plasmid during co-transfection.
  • the transiently transfected cells were assayed for luciferase activity 48 hours after plasmid transfection (supra).
  • the luciferase activity expressed from a plasmid containing no E-box or STAR/control elements was subtracted, and the luciferase activities were normalized to protein content as described (supra).
  • FIG. 14 shows the results of the enhancer-blocking assay.
  • the E47/E-box enhancer complex activates expression of luciferase (“vector”); this enhanced level of expression has been normalized to 100. Enhancer activity is blocked by all STAR elements tested. Enhancer activity is also blocked by the HS4 and scs elements, as expected (Bell et al., 2001, Gerasimova and Corces, 2001).
  • FIG. 15 shows the results of the enhancer assay.
  • the level of luciferase expression due to enhancement by the E47/E-box complex is set at 100 (“E47”).
  • E47 the level of luciferase expression due to enhancement by the E47/E-box complex
  • none of the STAR elements bring about significant activation of luciferase expression.
  • the scs and HS4 elements also do not bring about activation of the reporter gene. Therefore, it is concluded that at least the tested STAR elements do not possess enhancer properties.
  • STAR6 (SEQ ID NO:6) is between the SLC8A1 and HAAO genes in both the human and mouse genomes.
  • a cloned human STAR has a paralog within the human genome, and its ortholog has been identified in the mouse genome.
  • STAR3a is a fragment of the 15q11.2 region of human chromosome 15. This region is 96.9% identical (paralogous) with a DNA fragment at 5q33.3 on human chromosome 5, which is near the IL12B interleukin gene.
  • These human DNAs share approximately 80% identity with a fragment of the 11B2 region on mouse chromosome 11.
  • the 11B2 fragment is also near the (mouse) IL12B interleukin gene. Therefore, STAR3a and the mouse 11B2 fragment can be strictly defined as paralogs.
  • the region of mouse/human sequence conservation around STAR18 was recovered from human BAC clone RP11-387A1 by PCR amplification, in three fragments: the entire region (primers E93 (SEQ ID NO:171) and E94 (SEQ ID NO:172)), the leftward half (primers E93 (SEQ ID NO:171) and E92 (SEQ ID NO:170)), and the rightward half (primers E57 (SEQ ID NO:169) and E94 (SEQ ID NO:172)).
  • the corresponding fragments from the homologous mouse region were recovered from BAC clone RP23-400H17 in the same fashion (primers E95 (SEQ ID NO:173) and E98 (SEQ ID NO:176), E95 (SEQ ID NO:173) and E96 (SEQ ID NO:174), and E97 (SEQ ID NO:175) and E98 (SEQ ID NO:176), respectively). All fragments were cloned into the pSelect vector and transfected into a U-2 OS/Tet-Off/LexA-HP1 cell line (supra). Following transfection, hygromycin selection was carried out to select for transfected cells. The LexA-HP1 protein was induced by lowering the doxycycline concentration, and the ability of the transfected cells to withstand the antibiotic zeocin (a measure of STAR activity) was assessed by monitoring cell growth.
  • the original STAR18 clone was isolated from Sau3AI digested human DNA ligated into the pSelect vector on the basis of its ability to prevent silencing of a zeocin resistance gene. Alignment of the human STAR18 clone (497 base pairs) with the mouse genome revealed high sequence similarity (72%) between the orthologous human and mouse STAR18 (SEQ ID NO:18) regions. It also uncovered high similarity (73%) in the region extending for 488 base pairs immediately to the left of the Sau3AI site that defines the left end of the cloned region ( FIG. 16 ). Outside these regions the sequence similarity between human and mouse DNA drops below 60%.
  • both the human and the mouse STAR18 (SEQ ID NO:18) elements confer survival on zeocin to host cells expressing the lexA-HP1 repressor protein.
  • the original 497 base pair STAR18 clone and its mouse ortholog both confer the ability to grow ( FIG. 16 , a and d ).
  • the adjacent 488 base pair regions of high similarity from both genomes also confer the ability to grow, and in fact their growth phenotype is more vigorous than that of the original STAR18 clone ( FIG. 16 , b and e ).
  • FIG. 16 , b and e When the entire region of sequence similarity was tested, these DNAs from both mouse and human confer growth, and the growth phenotype is more vigorous than the two sub-fragments ( FIG.
  • STAR elements are isolated on the basis of their anti-repression phenotype with respect to transgene expression.
  • This anti-repression phenotype reflects underlying biochemical processes that regulate chromatin formation which are associated with the STAR elements. These processes are typically sequence-specific and result from protein binding or DNA structure. This suggests that STAR elements will share DNA sequence similarity. Identification of sequence similarity among STAR elements will provide sequence motifs that are characteristic of the elements that have already been identified by functional screens and tests. The sequence motifs will also be useful to recognize and claim new STAR elements whose functions conform to the claims of this patent. The functions include improved yield and stability of transgenes expressed in eukaryotic host cells.
  • sequence motifs that characterize STAR elements include: (1) provision of search motifs for prediction and identification of new STAR elements in genome databases, (2) provision of a rationale for modification of the elements, and (3) provision of information for functional analysis of STAR activity. Using bio-informatics, sequence similarities among STAR elements have been identified; the results are presented in this example.
  • Regulatory DNA elements typically function via interaction with sequence-specific DNA-binding proteins.
  • Bio-informatic analysis of DNA elements such as STAR elements whose regulatory properties have been identified, but whose interacting proteins are unknown, requires a statistical approach for identification of sequence motifs. This can be achieved by a method that detects short DNA sequence patterns that are over-represented in a set of regulatory DNA elements (e.g., the STAR elements) compared to a reference sequence (e.g., the complete human genome). The method determines the number of observed and expected occurrences of the patterns in each regulatory element. The number of expected occurrences is calculated from the number of observed occurrences of each pattern in the reference sequence.
  • the DNA sequence patterns can be oligonucleotides of a given length, e.g., six base pairs.
  • the actual frequency of each hexamer in the reference sequence is typically different than this due to biases in the content of G:C base pairs, etc. Therefore, the frequency of each oligonucleotide in the reference sequence is determined empirically by counting, to create a “frequency table” for the patterns.
  • the pattern frequency table of the reference sequence is then used to calculate the expected frequency of occurrence of each pattern in the regulatory element set.
  • the expected frequencies are compared with the observed frequencies of occurrence of the patterns. Patterns that are “over-represented” in the set are identified; for example, if the hexamer ACGTGA (SEQ ID NO:123) is expected to occur five times in 20 kilobase pairs of sequence, but is observed to occur 15 times, then it is three-fold over-represented. Ten of the 15 occurrences of that hexameric sequence pattern would not be expected in the regulatory elements if the elements had the same hexamer composition as the entire genome.
  • a significance index, “sig,” is calculated for each pattern.
  • the significance index is derived from the probability of occurrence of each pattern, which is estimated by a binomial distribution. The probability takes into account the number of possible patterns (4096 for hexamers). The highest sig values correspond to the most overrepresented oligonucleotides (van Helden et al., 1998). In practical terms, oligonucleotides with sig ⁇ 0 are considered as over-represented.
  • the patterns that are significantly over-represented in the regulatory element set are used to develop a model for classification and prediction of regulatory element sequences.
  • Discriminant Analysis sets of known or classified items (e.g., STAR elements) are used to “train” a model to recognize those items on the basis of specific variables (e.g., sequence patterns such as hexamers).
  • the trained model is then used to predict whether other items should be classified as belonging to the set of known items (e.g., is a DNA sequence a STAR element).
  • the known items in the training set are STAR elements (positive training set).
  • Discriminant Analysis establishes criteria for discriminating positives from negatives based on a set of variables that distinguish the positives; in this example, the variables are the significantly over-represented patterns (e.g., hexamers).
  • Over-training is circumvented by applying a forward stepwise selection of variables (Huberty, 1994).
  • the goal of Stepwise Discriminant Analysis is to select the minimum number of variables that provides maximum discrimination between the positives and negatives.
  • the model is trained by evaluating variables one-by-one for their ability to properly classify the items in the positive and negative training sets. This is done until addition of new variables to the model does not significantly increase the model's predictive power (i.e., until the classification error rate is minimized). This optimized model is then used for testing, in order to predict whether “new” items are positives or negatives (Huberty, 1994).
  • the first step reduces a large set of sequence patterns (e.g., all 4096 hexamers) to a smaller set of significantly over-represented patterns (e.g., 100 hexamers); in the second step, Stepwise Discriminant Analysis reduces the set of over-represented patterns to the subset of those patterns that have maximal discriminative power (e.g., five to ten hexamers). Therefore, this approach provides simple and robust criteria for identifying regulatory DNA elements such as STAR elements.
  • DNA-binding proteins can be distinguished on the basis of the type of binding site they occupy. Some recognize contiguous sequences; for this type of protein, patterns that are oligonucleotides of length six base pairs (hexamers) are fruitful for bio-informatic analysis (van Helden et al., 1998). Other proteins bind to sequence dyads: contact is made between pairs of highly conserved trinucleotides separated by a non-conserved region of fixed width (van Helden et al., 2000).
  • BAC/PAC bacterial/P1
  • the BAC/PAC clones contain genomic DNA from regions of chromosome 1 (clones RP1154H19 and RP3328E19), from the HOX cluster of homeotic genes (clones RP1167F23, RP1170019, and RP11387A1), or from human chromosome 22 (Research Genetics 96010-22).
  • the DNAs were size-fractionated, and the 0.5-2 kb size fraction was ligated into BamHI-digested pSelect vector, by standard techniques (Sambrook et al., 1989).
  • pSelect plasmids containing human genomic DNA that conferred resistance to zeocin at low doxycycline concentrations were isolated and propagated in Escherichia coli .
  • the screens that yielded the STAR elements of Table 3 have assayed approximately 1-2% of the human genome.
  • the human genomic DNA inserts in these 66 plasmids were sequenced by the dideoxy method (Sanger et al., 1977) using a Beckman CEQTM2000 automated DNA sequencer, using the manufacturer's instructions. Briefly, DNA was purified from E. coli using QIAprep® Spin Miniprep and Plasmid Midi Kits (QIAGEN® 27106 and 12145, respectively). Cycle sequencing was carried out using custom oligonucleotides corresponding to the pSelect vector (primers D89 (SEQ ID NO:149) and D95 (SEQ ID NO:154), Table 2), in the presence of dye terminators (CEQTM Dye Terminator Cycle Sequencing Kit, Beckman 608000).
  • Assembled STAR DNA sequences were located in the human genome (database builds August and December 2001) using BLAT (Basic Local Alignment Tool (Kent, 2002); http://genome.ucsc.edu/cgi-bin/hgGateway; Table 3).
  • the combined STAR sequences comprise 85.6 kilobase pairs, with an average length of 1.3 kilobase pairs.
  • Sequence motifs that distinguish STAR elements within human genomic DNA were identified by bio-informatic analysis using a two-step procedure, as follows (see FIG. 17 for a schematic diagram).
  • the analysis has two input datasets: (1) the DNA sequences of the STAR elements (STAR1-STAR65 (SEQ ID NOS:1-65) were used; Table 3); and (2) the DNA sequence of the human genome (except for chromosome 1, which was not feasible to include due to its large size; for dyad analysis a random subset of human genomic DNA sequence ( ⁇ 27 Mb) was used).
  • the first step in the analysis uses RSA-Tools software (Regulatory Sequence Analysis Tools; http://www.ucmb.ulb.ac.be/bioinformatics/rsa-tools/; references (van Helden et al., 1998, van Helden et al., 2000, van Helden et al., 2000)) to determine the following information: (1) the frequencies of all dyads and hexameric oligonucleotides in the human genome; (2) the frequencies of the oligonucleotides and dyads in the 65 STAR elements; and (3) the significance indices of those oligonucleotides and dyads that are over-represented in the STAR elements compared to the genome.
  • a control analysis was done with 65 sequences that were selected at random from the human genome (i.e., from 2689 ⁇ 10 3 kilobase pairs) that match the length of the STAR elements of Table 3.
  • the over-represented oligonucleotides and dyads were used to train models for prediction of STAR elements by Linear Discriminant Analysis (Huberty, 1994).
  • a pre-selection of variables was performed by selecting the 50 patterns with the highest individual discriminatory power from the over-represented oligos or dyads of the frequency analyses. These pre-selected variables were then used for model training in a Stepwise Linear Discriminant Analysis to select the most discriminant combination of variables (Huberty, 1994).
  • Variable selection was based on minimizing the classification error rate (percentage of false negative classifications).
  • the expected error rate was estimated by applying the same discriminant approach to the control set of random sequences (minimizing the percentage of false positive classifications).
  • the predictive models from the training phase of Discriminant Analysis were tested in two ways. First, the STAR elements and random sequences that were used to generate the model (the training sets) were classified. Second, sequences in a collection of 19 candidate STAR elements (recently cloned by zeocin selection as described above) were classified. These candidate STAR elements are listed in Table 8 (SEQ ID NOS:66-84).
  • Pattern frequency analysis was performed with RSA-Tools on 65 STAR elements, using the human genome as the reference sequence.
  • One hundred sixty-six (166) hexameric oligonucleotides were found to be over-represented in the set of STAR elements (sig ⁇ 0) compared to the entire genome (Table 6).
  • the most significantly over-represented oligonucleotide, CCCCAC (SEQ ID NO:177) occurs 107 times among the 65 STAR elements, but is expected to occur only 49 times. It has a significance coefficient of 8.76; in other words, the probability that its over-representation is due to random chance is 1/10 8.76 , i.e., less than one in 500 million.
  • Table 6 also presents the number of STAR elements in which each over-represented oligonucleotide is found.
  • oligo 1 CCCCAC (SEQ ID NO:177)
  • oligo 1 CCCCAC (SEQ ID NO:177)
  • AATCGG SEQ ID NO:342
  • single-copy oligonucleotides occur frequently, especially for the lower-abundance oligos.
  • oligo 4 occurs on average three times in those STARs in which it is found (37 STARs).
  • the most widespread oligonucleotide is number 120 (CCCAGG (SEQ ID NO:296)), which occurs on 58 STARs (on average twice per STAR), and the least widespread oligonucleotide is number 114 (CGTCGC (SEQ ID NO:290)), which occurs on only six STARs (and on average only once per STAR).
  • Results of dyad frequency analysis are given in Table 7. Seven hundred thirty (730) dyads were found to be over-represented in the set of STAR elements (sig ⁇ 0) compared to the reference sequence. The most significantly over-represented dyad, CCCN ⁇ 2 ⁇ CGG (SEQ ID NO:343), occurs 36 times among the 65 STAR elements, but is expected to occur only seven times. It has a significance coefficient of 9.31; in other words, the probability that its over-representation is due to chance is 1/10 9.31 , i.e., less than one in two billion.
  • Three hundred ninety-seven (397) of the dyads have a significance coefficient greater than 1, and are, therefore, highly over-represented in the STAR elements.
  • oligonucleotides and dyads found to be over-represented in STAR elements by pattern frequency analysis were tested for their discriminative power by Linear Discriminant Analysis.
  • Discriminant models were trained by step-wise selection of the best combination among the 50 most discriminant oligonucleotide (Table 6) or dyad (Table 7) patterns. The models achieved optimal error rates after incorporation of four (dyad) or five variables.
  • the discriminative variables from oligo analysis are numbers 11, 30, 94, 122, and 160 (Table 6); those from dyad analysis are numbers 73, 194, 419, and 497 (Table 7).
  • the discriminant models were then used to classify the 65 STAR elements in the training set and their associated random sequences.
  • the model using oligonucleotide variables classifies 46 of the 65 STAR elements as STAR elements (true positives); the dyad model classifies 49 of the STAR elements as true positives.
  • the models classify 59 of the 65 STAR elements as STAR elements (91%; FIG. 18 ).
  • the false positive rates random sequences classified as STARs were seven for the dyad model, eight for the oligonucleotide model, and 13 for the combined predictions of the two models (20%).
  • the STAR elements of Table 3 that were not classified as STARs by LDA are STAR7, STAR22, STAR35, STAR44, STAR46, and STAR65 (SEQ ID NOS:7, 22, 35, 44, 46 and 65, respectively). These elements display stabilizing anti-repressor activity in functional assays, so the fact that they are not classified as STARs by LDA suggests that they represent another class (or classes) of STAR elements.
  • the models were then used to classify the 19 candidate STAR elements in the testing set listed in Table 8.
  • the dyad model classifies 12 of these candidate STARs as STAR elements, and the oligonucleotide model classifies 14 as STARs.
  • the combined number of the candidates that are classified as STAR elements is 15 (79%). This is a lower rate of classification than obtained with the training set of 65 STARs; this is expected for two reasons.
  • the discriminant models were trained with the 65 STARs of Table 3, and discriminative variables based on this training set may be less well represented in the testing set.
  • the candidate STAR sequences in the testing set have not yet been fully characterized in terms of in vivo function, and may include elements with only weak anti-repression properties.
  • the STAR sequences contain a number of dyad and hexameric oligonucleotide patterns that are significantly over-represented in comparison with the human genome as a whole. These patterns may represent binding sites for proteins that confer STAR activity; in any case they form a set of sequence motifs that can be used to recognize STAR element sequences.
  • Transgene silencing occurs in transgenic plants at both the transcriptional and post-transcriptional levels (Meyer, 2000, Vance and Vaucheret, 2001). In either case, the desired result of transgene expression can be compromised by silencing; the low expression and instability of the transgene results in poor expression of desirable traits (e.g., pest resistance) or low yields of recombinant proteins. It also results in poor predictability: the proportion of transgenic plants that express the transgene at biotechnologically useful levels is low, which necessitates laborious and expensive screening of transformed individuals for those with beneficial expression characteristics.
  • This example describes the isolation of STAR elements from the genome of the dicot plant Arabidopsis thaliana for use in preventing transcriptional transgene silencing in transgenic plants.
  • Arabidopsis was chosen for this example because it is a well-studied model organism: it has a compact genome, it is amenable to genetic and recombinant DNA manipulations, and its genome has been sequenced (Bevan et al., 2001, Initiative, 2000, Meinke et al., 1998).
  • Genomic DNA was isolated from Arabidopsis thaliana ecotype Columbia as described (Stam et al., 1998) and partially digested with MboI. The digested DNA was size-fractionated to 0.5-2 kilobase pairs by agarose gel electrophoresis and purification from the gel (QIAquick® Gel Extraction Kit, QIAGEN® 28706), followed by ligation into the pSelect vector (supra). Transfection into the U-2 OS/Tet-Off/LexA-HP1 cell line and selection for zeocin resistance at low doxycycline concentration was performed as described (supra). Plasmids were isolated from zeocin resistant colonies and re-transfected into the U-2 OS/Tet-Off/LexA-HP1 cell line.
  • STAR activity was tested further by measuring mRNA levels for the hygromycin- and zeocin-resistance genes in recombinant host cells by reverse transcription PCR (RT-PCR).
  • RT-PCR reverse transcription PCR
  • Cells of the U-2 OS/Tet-Off/lexA-HP1 cell line were transfected with pSelect plasmids containing Arabidopsis STAR elements, the Drosophila scs element, or containing no insert (supra). These were cultivated on hygromycin for two weeks at high doxycycline concentration, then the doxycycline concentration was lowered to 0.1 ng/ml to induce the lexA-HP1 repressor protein.
  • First-strand cDNA synthesis was carried out using the RevertAidTM First Strand cDNA Synthesis kit (MBI Fermentas 1622) using oligo(dT)18 primer as described by the manufacturer.
  • An aliquot of the cDNA was used as the template in a PCR reaction using primers D58 (SEQ ID NO:145) and D80 (SEQ ID NO:148) (for the zeocin marker), and D70 (SEQ ID NO:146) and D71 (SEQ ID NO:147) (for the hygromycin marker), and Taq DNA polymerase (Promega M2661).
  • the reaction conditions were 15-20 cycles of 94° C. for one minute, 54° C. for one minute, and 72° C. for 90 seconds. These conditions result in a linear relationship between input RNA and PCR product DNA.
  • the PCR products were resolved by agarose gel electrophoresis, and the zeocin and hygromycin bands were detected by Southern blotting as described (Sambrook et al., 1989), using PCR products produced as above with purified pSelect plasmid as template. The ratio of the zeocin and hygromycin signals corresponds to the normalized expression level of the zeocin gene.
  • the library of Arabidopsis genomic DNA in the pSelect vector comprised 69,000 primary clones in E. coli, 80% of which carried inserts.
  • the average insert size was approximately 1000 base pairs; the library, therefore, represents approximately 40% of the Arabidopsis genome.
  • the strength of the cloned Arabidopsis STAR elements was tested by assessing their ability to prevent transcriptional repression of the zeocin-resistance gene, using an RT-PCR assay. As a control for RNA input among the samples, the transcript levels of the hygromycin-resistance gene for each STAR transfection were assessed too. This analysis has been performed for 12 of the Arabidopsis STAR elements. The results ( FIG. 19 ) demonstrate that the Arabidopsis STAR elements are superior to the Drosophila scs element (positive control) and the empty vector (“SV40”; negative control) in their ability to protect the zeocin-resistance gene from transcriptional repression.
  • STAR-A28 (SEQ ID NO:112) and STAR-A30 (SEQ ID NO:114) enable two-fold higher levels of zeocin-resistance gene expression than the scs element (normalized to the internal control of hygromycin-resistance gene mRNA) when the lexA-HP1 repressor is expressed.
  • STAR elements function to block the effect of transcriptional repression influences on transgene expression units.
  • Two of the benefits of STAR elements for heterologous protein production are an increased predictability to find high-expressing primary recombinant host cells as well as increased protein production or yield in these cells.
  • the disclosed STAR elements are human DNA sequences, isolated in the human U-2 OS osteosarcoma cell line. It is, therefore, an important question whether the human STAR elements are functional in a) cell lines derived from species other than man, and/or in b) human cell lines other than the U-2 OS osteosarcoma cell line.
  • the functionality of STAR 7 (SEQ ID NO:7) in (CHO) Chinese hamster ovary are illustrated.
  • the STAR7 (SEQ ID NO:7) element is tested in the ppGIZ-STAR7 vector ( FIG. 20 ).
  • the construction of the pPlug&Play-GFP-ires-Zeo (ppGIZ) vector is described below.
  • Plasmid pGFP (Clontech 6010-1) is modified by insertion of a linker at the BsiWI site to yield pGFP-link.
  • the linker (made by annealing oligonucleotides 5′ GTACGGATATCAGATCTTTAATTAAG 3′ (SEQ ID NO:124) and 5′ GTACCTTAATTAAAGATCTGATATCC 3′ (SEQ ID NO:125)) introduces sites for the PacI, BglII, and EcoRV restriction endonucleases. This creates the multiple cloning site MCSII for insertion of STAR elements.
  • primers 5′ ATCAGATCTGGCGCGCCATTTAAATCGTC TCGCGCGTTTCGGTGATGACGG 3′ (SEQ ID NO:126) and 5′ AGGCGGATCCGAATG TATTTAGAAAAATAAACAAATAGGGG 3′ (SEQ ID NO:127) are used to amplify a region of 0.37 kb from pGFP, which is inserted into the BglII site of pIRES (Clontech 6028-1) to yield pIRES-stuf.
  • pIRES-stuf is digested with BglII and FspI to liberate a DNA fragment composed of the stuffer fragment, the CMV promoter, the IRES element (flanked by multiple cloning sites MCS A and MCS B), and the SV40 polyadenylation signal.
  • This fragment is ligated with the vector backbone of pGFP-link produced by digestion with BamHI and StuI, to yield pIRES-link.
  • the open reading frames of the zeocin-resistance gene is inserted into the BamHI/NotI sites of MCS B in pIRES-link as follows: the zeocin-resistance ORF is amplified by PCR with primers 5′ GATCGGATCCTTCGAAATGGCCAAGTTGACCAGTGC 3′ (SEQ ID NO:128) and 5′ AGGCGCGGCCGCAATTCTCAGTCCTGCTCCTC 3′ (SEQ ID NO:129) from plasmid pEM7/zeo, digested with BamHI and NotI, and ligated with BamHI/NotI-digested pIRES-link to yield pIRES-link-zeo.
  • the GFP reporter ORF was introduced into pIRES-link-zeo by amplification of phr-GFP-1 with primers 5′ GATCGAATTCTCGCGAATGGTGAGCAAGCAGATCCTGAAG 3′ (SEQ ID NO:130) and 5′ AGGCGAATTCACCGGTGTTTAAACTTACACCCACTCGTGCAGGCTGCCCAGG 3′ (SEQ ID NO:131), and insertion of the EcoRI-digested GFP cassette into the EcoRI site in MCS A of the pIRES-link-zeo plasmid. This created the ppGIZ (for ppGFP-IRES-zeo). STAR7 (SEQ ID NO:7) is cloned into the SalI site (5′) and into the PacI site (3′).
  • the Chinese Hamster Ovary cell line CHO-K1 (ATCC CCL-61) is cultured in HAMS-F12 medium+10% Fetal Calf Serum containing 2 mM glutamine, 100 U/ml penicillin, and 100 micrograms/ml streptomycin at 37° C./5% CO 2 .
  • Cells are transfected with the plasmids using Lipofectamine 2000 (Invitrogen) as described by the manufacturer. Briefly, cells are seeded to culture vessels and grown overnight to 70-90% confluence.
  • Lipofectamine reagent is combined with plasmid DNA at a ratio of 7.5 microliters per 3 microgram (e.g., for a 10 cm Petri dish, 20 micrograms DNA and 120 microliters Lipofectamine) and added after a 30-minute incubation at 25° C. to the cells. After a six-hour incubation, the transfection mixture is replaced with fresh medium, and the transfected cells are incubated further. After overnight cultivation, cells are trypsinized and seeded into fresh petri dishes with fresh medium with zeocin added to a concentration of 100 ⁇ g/ml and the cells are cultured further.
  • the tested constructs consist of a bicistronic gene with the GFP gene, an IRES and the Zeocin resistance gene under control of the CMV promoter, but either with or without STAR7 (SEQ ID NO:7) element to flank the entire construct ( FIG. 20 ).
  • the constructs are transfected to CHO-K1 cells. Stable colonies are expanded before the GFP signal is determined on a XL-MCL Beckman Coulter flow cytometer. The mean of the GFP signal is taken as measure for the level of GFP expression and this is plotted in FIG. 20 .
  • FIG. 20 shows that flanking a GFP reporter gene that is under the control of the CMV promoter results in a higher number of CHO colonies that express significantly higher levels of GFP protein, as compared to the control without STAR7 (SEQ ID NO:7) element.
  • the STAR7 (SEQ ID NO:7) element therefore, conveys a higher degree of predictability of transgene expression in CHO cells.
  • the highest GFP expression level in STAR-shielded CHO colonies is also higher than in STAR-less control colonies.
  • STAR elements function to block the effect of transcriptional repression influences on transgene expression units.
  • Two of the benefits of STAR elements for heterologous protein production are an increased predictability to find high-expressing primary recombinant host cells, as well as increased protein production or yield in these cells.
  • the disclosed STAR elements are human DNA sequences, isolated in the human U-2 OS osteosarcoma cell line. It is, therefore, an important question whether the human STAR elements are functional in a) cell lines derived from species other than man, and/or in b) human cell lines other than the U-2 OS osteosarcoma cell line.
  • STAR 7 SEQ ID NO:7 in non-secreting mouse myeloma (NSO) cells are illustrated.
  • NSO (Non-Secreting mouse myeloma) cells (ECACC 85110503) are suspension cells that are cultured in RPMI 1640 medium+10% Fetal Calf Serum containing 2 mM glutamine, 100 U/ml penicillin, and 100 micrograms/ml streptomycin at 37° C./5% CO 2 .
  • Cells are transfected with the plasmids using Lipofectamine 2000 (Invitrogen) as described by the manufacturer. Briefly, cells are seeded to culture vessels and grown overnight to 4 ⁇ 10 5 /ml.
  • Lipofectamine reagent is combined with plasmid DNA at a ratio of 3 microliters per microgram DNA (e.g., for a 10 cm Petri dish, 20 micrograms DNA and 60 microliters Lipofectamine) and added after 30 minutes incubation at 25° C. temperature to the cells. After overnight incubation, the transfection mixture is replaced with fresh medium and the transfected cells are incubated further. After another overnight incubation, zeocin is added to a concentration of 100 ⁇ g/ml and the cells are cultured and further incubated for three days. Then the cells are seeded in 96-well plates in such dilutions that one well will contain ⁇ 1 cell. After ten days growing colonies are transferred to 24-well plates.
  • 3 microliters per microgram DNA e.g., for a 10 cm Petri dish, 20 micrograms DNA and 60 microliters Lipofectamine
  • FIG. 21 shows that flanking a GFP reporter gene that is under the control of the CMV promoter results in a higher number of NSO colonies that express significantly higher levels of GFP protein, as compared to the control without STAR7 (SEQ ID NO:7) element.
  • the STAR7 (SEQ ID NO:7) element therefore, conveys a higher degree of predictability of transgene expression in NSO cells.
  • the highest GFP expression level in STAR-shielded NSO colonies is also higher than in STAR-less control colonies. It is, therefore, concluded that STAR7 (SEQ ID NO:7) is able to convey higher expression levels to a transgene in NSO cells, this being a cell line derived from another species than man.
  • STAR elements function to block the effect of transcriptional repression influences on transgene expression units.
  • Two of the benefits of STAR elements for heterologous protein production are an increased predictability to find high-expressing primary recombinant host cells as well as increased protein production or yield in these cells.
  • the disclosed STAR elements are human DNA sequences, isolated in the human U-2 OS osteosarcoma cell line. It is, therefore, an important question whether the human STAR elements are functional in a) cell lines derived from species other than man, and/or in b) human cell lines other than the U-2 OS osteosarcoma cell line.
  • the functionality of STAR7 (SEQ ID NO:7) in human 293 cells are illustrated.
  • the tested constructs are the same as described in Example 14.
  • the 293 cell line (ATCC CRL-1573) is derived from human embryonal kidney (immortalized by adenovirus 5 transfection) and is cultured in Dulbecco's Modified Eagle Medium+10% Fetal Calf Serum containing 2 mM glutamine, 100 U/ml penicillin, and 100 micrograms/ml streptomycin at 37° C./5% CO 2 . Cells are transfected with the plasmids using Lipofectamine 2000 (Invitrogen) as described by the manufacturer. Selection and propagation of the 293 colonies are as described in Example 14 for U-2 OS cells.
  • FIG. 22 shows that flanking a GFP reporter gene that is under the control of the CMV promoter results in a higher number of 293 colonies that express significantly higher levels of GFP protein, as compared to the control without STAR7 (SEQ ID NO:7) element.
  • the STAR7 (SEQ ID NO:7) element therefore, conveys a higher degree of predictability of transgene expression in 293 cells.
  • the highest GFP expression level in STAR-shielded colonies is also higher than in STAR-less control colonies. It is, therefore, concluded that STAR7 (SEQ ID NO:7) is able to convey higher expression levels to a transgene in 293 cells, this being another human cell line, distinct from the human U-2 OS cell line.
  • cytogenetic ideogram of each chromosome e.g., 1p2.3 is the third cytogenetic sub-band of the second cytogenetic band of the short arm of chromosome 1 (http://www.ncbi.nlm.nih.gov/Class/MLACourse/Genetics/chrombanding.html).
  • F forward sequencing reaction result
  • R reverse sequencing reaction result.
  • STAR elements convey stability over time on transgene expression 1 Cell Divisions 2 Luciferase Expression 3 STAR6 (SEQ ID NO: 6) 42 18,000 plus puromycin 60 23,000 84 20,000 108 16,000 STAR6 (SEQ ID NO: 6) 84 12,000 without puromycin 4 108 15,000 144 12,000 1 Plasmid pSDH-Tet-STAR6 was transfected into U-2 OS cells, and clones were isolated and cultivated in doxycycline-free medium as described in Example 1. Cells were transferred to fresh culture vessels weekly at a dilution of 1:20. 2 The number of cell divisions is based on the estimation that in one week the culture reaches cell confluence, which represents ⁇ 6 cell divisions. 3 Luciferase was assayed as described in Example 1. 4 After 60 cell divisions the cells were transferred to two culture vessels; one was supplied with culture medium that contained puromycin, as for the first 60 cell divisions, and the second was supplied with culture medium lacking antibiotic.
  • the patterns are ranked according to significance coefficient. These were determined using RSA-Tools with the sequence of the human genome as reference. Patterns that comprise the most discriminant variables in Linear Discriminant Analysis are indicated with an asterisk. (SEQ ID NOS:177-342)
  • the patterns are ranked according to significance coefficient. These were determined using RSA-Tools with the random sequence from the human genome as reference. Patterns that comprise the most discriminant variables in Linear Discriminant Analysis are indicated with an asterisk. (SEQ ID NOS:343-1072)
  • Arabidopsis STAR elements of the invention including chromosome location and length (SEQ ID NOS: 85-119) STAR Chromosome Length, kb SEQ ID NO: A1 I 1.2 85 A2 I 0.9 86 A3 I 0.9 87 A4 I 0.8 88 A5 I 1.3 89 A6 I 1.4 90 A7 II 1.2 91 A8 II 0.8 92 A9 II 0.9 93 A10 II 1.7 94 A11 II 1.9 95 A12 II 1.4 96 A13 II 1.2 97 A14 II 2.1 98 A15 II 1.4 99 A16 II 0.7 100 A17 II 1.5 101 A18 III 1.5 102 A19 III 0.7 103 A20 III 2.0 104 A21 IV 1.8 105 A22 IV 0.8 106 A23 IV 0.6 107 A24 IV 0.5 108 A25 V 0.9 109 A26 V 1.9 110 A27 V 1.1 111 A28 V 1.6 112 A29 V 0.9 113 A30 V 2.0 114 A31 V 2.0

Abstract

The invention relates to means and methods for regulating gene expression and production of proteinaceous molecules. The invention provides a method for producing a proteinaceous molecule in a cell comprising selecting a cell for its suitability for producing the proteinaceous molecule, providing a nucleic acid encoding the proteinaceous molecule with a nucleic acid comprising a STAR (STabilizing Anti-Repression) sequence, expressing the resulting nucleic acid in the cell and collecting the proteinaceous molecule. Providing at least one STAR sequence to a nucleic acid encoding a proteinaceous molecule will enhance production (yield) of the proteinaceous molecule by a host cell, increase the proportion of host cells with acceptable expression levels, and/or increase stability of a gene expression level.

Description

CROSS-REFERENCE TO RELATED APPLICATIONS
This application is a divisional of U.S. patent application Ser. No. 11/012,546, filed Dec. 14, 2004, now U.S. Pat. No. 7,267,965, which application is a continuation of International Patent Application No. PCT/NL03/00410, filed May 30, 2003, published in English as International Patent Publication No. WO 03/106674 on Dec. 24, 2003, which claims the benefit under 35 U.S.C. §119 of European Patent Application No. EP 02077344.6, filed Jun. 14, 2002, the entirety of each of which are hereby incorporated by reference.
STATEMENT ACCORDING TO 37 C.F.R §1.52(e)(5)—SEQUENCE LISTING SUBMITTED ON COMPACT DISC
Pursuant to 37 C.F.R. §1.52(e)(1)(iii), a compact disc containing an electronic version of the Sequence Listing has been submitted concomitant with this application, the contents of which are hereby incorporated by reference. The compact disc contains the file “P60555PC00 final.txt” which is 496 KB, and created on Dec. 14, 2004. A second compact disk is submitted and is an identical copy of the first compact disc (labeled, “copy 1” and “copy 2,” respectively).
TECHNICAL FIELD
The invention relates to the fields of medicine and cellular biology. More specifically, the invention relates to means and methods for regulating gene expression, and production of proteinaceous molecules.
BACKGROUND OF THE INVENTION
Polypeptide production at industrial scale currently provides many biologically active polypeptides for a variety of uses, including diagnostic and therapeutic pharmaceuticals, industrial catalysts and nourishment. Polypeptides are produced in a variety of host systems, including transgenic animals and plants, microbes, and cultured mammalian cells. In most cases, the host system is modified by recombinant DNA techniques, for instance resulting in introduction into the host cell of a transgene which encodes a polypeptide of interest. Such a transgene typically includes elements that influence the transcription, translation, and/or processing of the transgene's polypeptide coding sequence. A recombinant host is then identified and isolated which has a suitable yield of a polypeptide of interest, and the cell population of this recombinant host is increased to an extent that it can produce the required amount of polypeptide.
The choice of the host system depends on a number of factors including: (1) the nature and intended use of a polypeptide, and (2) the cost of production. For production of biopharmaceuticals, e.g., therapeutic proteins such as hormones, cytokines, and antibodies, the host system of choice is usually cultured mammalian cells. Considerations with respect to product use and production cost with host cells will be discussed below.
(1) For in vivo therapeutic use, a therapeutic protein must not only have the correct biological activity to alter the course of a disease. It must also do no harm. Most therapeutic proteins are exported from the cell by the secretory pathway. Secreted proteins are modified by a series of post-translational events, including glycosylation, disulfide bond formation, and proteolytic processing. The post-translational modification systems vary among different species and cell types in their detailed mechanisms of action. As a result, the same polypeptide chain can be detectably different when it is produced in different host cells. These differences can be analytical, such as differences in physical properties such as molecular mass, net electrical charge, carbohydrate composition, or structure. The differences can also be functional, affecting for instance the biological activity of the protein itself (catalytic activity, ligand binding characteristics, etc.), and/or its in vivo properties (immunogenicity, biological half life, biodistribution, etc.). Functional differences can, therefore, affect both function and possible side effect(s) of a therapeutic protein. Host cell lines that produce proteins with low efficacy are not suitable for commercial exploitation. Furthermore, host cells which produce modified protein that involves significant side effects in a patient should not be used. These factors are becoming increasingly important considerations during selection of a host cell line for production of a therapeutic protein.
(2) Therapeutic protein production in host cells is an intrinsically costly process. Current methods for industrial production of such proteins often perform poorly, resulting in products that are prohibitively expensive. Poor performance can be due to limitations of protein expression systems and host cell lines currently in use. These limitations mostly have a few specific causes, including (a) failure to identify and isolate recombinant host cell lines that have suitable productivity of proteins (poor predictability), (b) silencing, during the industrial production cycle, of the transgenes that encode proteins (poor stability), and (c) low or incorrect post-translational processing and secretory capacity of the host cell line. These limitations will be considered separately below.
(a) Conventional methods furnish only low frequencies of recombinant host cells that have suitable yields of proteins. Identifying and isolating these rare recombinant cell lines is a laborious and expensive process. The poor predictability of conventional methods means that often a recombinant host cell line is selected for production that has sub-optimal productivity characteristics, simply because a superior recombinant cell line was not identified and isolated during the selection process.
(b) Transgenes are often subject to silencing during cultivation of recombinant host cells. Silencing acts by suppressing transcription of a transgene. Detailed mechanisms of silencing are still not known, and different conventional methods are prone to different kinds of silencing phenomena. With one phenomenon, an individual transgene is silenced by formation of transcriptionally refractory heterochromatin at the transgenic locus. Heterochromatin formation is influenced by the position of genomic integration of a transgene (“position effects” (Boivin and Dura, 1998)). Transgene integration occurs more or less at random. Since most of the genome is heterochromatin, most transgene loci are prone to silencing due to position effects.
A second transgene-silencing phenomenon can occur when two or more copies of a transgene are integrated into a genome during construction of a recombinant cell line. Formation of tandem transgene repeats often occurs during the initial integration step. Furthermore, in order to increase product yield, many recombinant host cell lines are engineered after the integration step to amplify the copy number of a transgene, which also results in tandem transgene repeats (Kaufman, 1990). Tandem repeats and other configurations of multiple transgene copies are particularly prone to silencing (“repeat-induced gene silencing” (Garrick et al., 1998)).
In case that a genome contains multiple copies of a transgene, the yield can also decline via another phenomenon than transcriptional silencing. The number of copies of the transgene can decline during cultivation of a recombinant host cell line. The productivity of such cell lines at the time of selection for use is correlated with a transgene copy number, and consequently as copies of a transgene are lost, the yield declines (Kaufman, 1990).
(c) Different cell types in a mammalian organism have different capacities for post-translational modification and secretion of proteins. The functions of some cell types include production of large quantities of secreted proteins; examples include lymphocytes (producing immunoglobulins), hepatocytes (producing serum proteins), and fibroblasts (producing extracellular matrix proteins). These cell types are favorable sources for deriving host cell lines for production of secreted heterologous proteins. More favorable is the use of a cell line whose progenitor organismal cell type secretes a protein or class of proteins of interest. For example, it is particularly favorable to express recombinant monoclonal antibodies in lymphocytes (or host cells derived from lymphocytes), erythropoietin in hepatocytes (or host cells derived from hepatocytes), and blood clotting factors (e.g., Factor VIII and van Willebrand's factor) in endothelial cells (or host cells derived from endothelial cells).
The use of specific cell types (or cell lines derived therefrom) for production of their affiliated proteins is favorable because such specific cell types will carry out proper post-translational modifications of produced proteins. However, specific cell types often do not have high secretory capacities. For example, cells of the central nervous system, such as neurons, have low intrinsic protein secretion capacities. These cells do secrete proteins, however, including neurotrophins. Neurotrophins regulate the fate and shape of neuronal cells during fetal and juvenile development. Moreover, they influence patterns of neuronal degeneration and regeneration in adults (Bibel and Barde, 2000). Production of neurotrophins for therapeutic applications has considerable biopharmaceutical value (e.g., Axokine™, recombinant ciliary neurotrophic factor from Regeneron). In order to produce heterologous neurotrophins with post-translational modifications (and hence functional properties) that match the naturally-occurring proteins, expression in host cells derived from the central nervous system is favorable. However, production of polypeptides such as neurotrophins in host cell lines such as those derived from neural tissue is inefficient using conventional methods. The predictability of identifying high-expressor isolates from these types of cell lines is often poor; the yield of proteins from such cell lines is generally low, and production levels are characteristically unstable.
Another drawback to a use of specific host cells for production of affiliated proteins is that it is usually difficult to isolate cell lines with favorable biotechnological characteristics. These characteristics for instance include the mode and rate of growth, and the ease of introduction of a transgene. Consequently, various general host cell lines have been established. Examples of these include CHO cells from Chinese hamster ovary (ATCC (American Type Culture Collection) CCL-61), BHK cells from baby hamster kidney (ATCC CCL-10), and Vero cells from African green monkey kidney (ATCC CCL-81). These “general purpose” host cell lines are widely used for production of a number of heterologous proteins. A disadvantage of general purpose cell lines is that the post-translational modifications of heterologous proteins produced by them often differ from the post-translational modifications of the naturally occurring proteins. These differences can have functional consequences resulting in side effects, as discussed above.
Table 1 lists a number of proteins that are currently in use or under development for biopharmaceutical applications. It also lists the tissue or cell type in which each protein is normally produced in the human body. These 24 proteins (or protein classes) come from a wide range of cells and tissue, ranging from highly secretory cells (hepatocytes, endothelial cells) to cells with low secretory capacity (e.g., neural tissue). Currently, neither general-purpose host cells nor specific host cells have qualities that enable optimal expression of the broad spectrum of biopharmaceutically important secreted proteins.
Hence, protein production by conventional host cell lines involves a lot of disadvantages and complications, for instance with respect to yield and post-translational modifications. There is a need in the art for improved protein production in recombinant host cell lines.
SUMMARY OF THE INVENTION
The present invention provides a method for producing a proteinaceous molecule in a cell comprising selecting a cell for its suitability for producing the proteinaceous molecule, providing a nucleic acid encoding the proteinaceous molecule with a nucleic acid comprising a STAR (STabilizing Anti-Repression) sequence, expressing the resulting nucleic acid in the cell and collecting the proteinaceous molecule.
The STAR sequence has to be operably linked to the nucleic acid encoding the proteinaceous molecule in order to be effective. In one embodiment of the invention, one STAR element is used. Preferably however, more than one STAR element is used. In a particularly preferred embodiment, the nucleic acid encoding the proteinaceous molecule is provided with two STAR sequences; one STAR sequence at the 5′ side of the coding sequence of the nucleic acid and one STAR sequence at the 3′ side of the coding sequence of the nucleic acid.
Description of STAR Elements
New transcription regulatory elements were disclosed by the present inventors, which are named STAR sequences (See EP 01202581.3). STAR sequences are nucleic acid sequences that comprise a capacity to influence transcription of genes in cis. Typically, although not necessarily, the STAR sequences do not code by themselves for a functional protein.
A STAR sequence has a gene transcription modulating quality in at least one type of cell. A STAR sequence is capable of enhancing gene transcription resulting in a higher yield, increasing the proportion of transgene-comprising host cells with acceptable expression levels, and/or increasing stability of transgenes in recombinant cell lines.
In EP 01202581.3 a method of detecting, and optionally selecting, a STAR sequence is provided, comprising providing a transcription system with a variety of a fragment-comprising vectors, the vectors comprising i) an element with a gene-transcription repressing quality, and ii) a promoter directing transcription of a reporter gene, the method further comprising performing a selection step in the transcription system in order to identify the STAR sequence. In a preferred embodiment, the fragments are located between i) the element with a gene-transcription repressing quality, and ii) the promoter directing transcription of the reporter gene. RNA polymerase initiates the transcription process after binding to a specific sequence, called the promoter, that signals where RNA synthesis should begin. A STAR sequence can enhance transcription from the promoter in cis, in a given cell type and/or a given promoter.
Methods disclosed in EP 01202581.3 have been used to isolate STAR elements from the human genome. Isolated human STAR elements have been placed in DNA vectors so as to flank transgene expression units, and the vectors have subsequently been integrated into host cell genomes. Transgene expression in these recombinant host cells has been compared to expression in similar host cells in which the expression units are not flanked by STAR elements. The results show that STAR elements have at least one of three consequences for production of (heterologous) proteinaceous molecule (also referred to as (heterologous) protein): (1) they increase the predictability of identifying host cell lines that express a proteinaceous molecule at industrially acceptable levels; (2) they result in host cell lines with increased protein yields; and/or (3) they result in host cell lines that exhibit more stable protein production during prolonged cultivation. Each of these attributes is discussed in more detail below:
(1) Increased predictability: Integration of transgene expression units can occur at random positions throughout the host cell genome. However, much of the genome is transcriptionally silent heterochromatin. When the expression units include STAR elements flanking the transgene, the position of integration has a reduced effect on expression. The STAR elements impair the ability of adjacent heterochromatin to silence the transgene. Consequently, the proportion of transgene-containing host cells with acceptable expression levels is increased.
(2) Yield: The levels of protein expression in primary populations of recombinant host cells, directly after transgene integration, have been surveyed. The expression level of individuals in the populations varies. However, when the transgenes are protected by STAR elements, the variability is reduced. This reduced variability is most conspicuous in that fewer clones are recovered that have low levels of expression. Furthermore, the populations with STAR elements commonly have individuals with strikingly high expression. These high-yielding individuals are favorable for production of proteinaceous molecules.
(3) Increased stability: STAR elements increase the stability of transgenes in recombinant host cell lines by ensuring that the transgenes are not transcriptionally silenced during prolonged cultivation. Comparative trials show that, under conditions in which transgenes that are not protected by STAR elements are progressively silenced (5 to 25 passages in cultivation), STAR element-protected transgenes continue to be expressed at high levels. This is an advantage during industrial production of proteinaceous molecules, during which cell cultivation continues for prolonged periods, from a few weeks to many months.
Hence, a STAR sequence can enhance expression of a heterologous proteinaceous molecule. In addition, a STAR sequence can enhance expression of a naturally produced proteinaceous molecule.
Transcription can be influenced through a direct effect of the STAR sequence (or the protein(s) binding to it) on the transcription of a particular promoter. Transcription can however, also be influenced by an indirect effect, for instance because the STAR sequence affects the function of one or more other regulatory elements. A STAR sequence can also comprise a stable gene transcription quality. Frequently, expression levels drop dramatically with increasing numbers of cell divisions. With the methods disclosed in EP 01202581.3 it is possible to detect and optionally select a DNA sequence that is capable of at least in part preventing the dramatic drop in transcription levels with increasing numbers of cell divisions. Strikingly, fragments comprising a STAR sequence can be detected and optionally selected with a method EP 01202581.3, in spite of the fact that the method does not necessarily measure long term stability of transcription.
A STAR sequence is suitable for enhancing the level of transcription of a gene of interest in a host cell. If, together with a gene of interest, a STAR sequence is also introduced into host cells, more clones can be detected that express more than a certain amount of the gene of interest. As used herein, such host cells are termed “host cells with acceptable expression levels.”
Furthermore, if, together with a gene of interest, a STAR sequence is also introduced into host cells, a higher yield of produced proteinaceous molecules can be obtained, while gene expression level is also more stable than in the absence of such STAR sequences. Preferably, a STAR sequence derived from a plant and/or vertebrate is used. More preferably a human STAR sequence is used.
Sequences comprising a STAR sequence can be found by using a functional assay, as described above. However, once a collection of such sequences has been identified, bioinformatics can be used to find other STAR sequences. Several methods are available in the art to extract sequence identifiers from a family of DNA sequences sharing a certain common feature. Such sequence identifiers can subsequently be used to identify sequences that share one or more identifiers. Sequences sharing such one or more identifiers are likely to be a member of the same family of sequences, i.e., are likely to share the common feature of the family. By the present inventors a large number of sequences comprising STAR activity (so-called STAR sequences) were used to obtain sequence identifiers (patterns) which are characteristic for sequences comprising STAR activity. These patterns can be used to determine whether a test sequence is likely to contain STAR activity. A method for detecting the presence of a STAR sequence within a nucleic acid sequence of about 50-5000 base pairs is thus provided, comprising determining the frequency of occurrence in the sequence of at least one sequence pattern and determining that the frequency of occurrence is representative of the frequency of occurrence of at least one sequence pattern in at least one sequence comprising a STAR sequence. In principle any method is suited for determining whether a sequence pattern is representative of a STAR sequence. Many different methods are available in the art. Preferably, the step of determining that the occurrence is representative of the frequency of occurrence of at least one sequence pattern in at least one sequence comprising a STAR sequence comprises, determining that the frequency of occurrence of at least one sequence pattern significantly differs between at least one STAR sequence and at least one control sequence. In principle any significant difference is discriminative for the presence of a STAR sequence. However, in a particularly preferred embodiment, the frequency of occurrence of at least one sequence pattern is significantly higher in at least one sequence comprising a STAR sequence compared to at least one control sequence.
A considerable number of sequences comprising a STAR sequence have been identified by the present inventors. It is possible to use these sequences to test how efficient a pattern is in discriminating between a control sequence and a sequence comprising a STAR sequence. Using so-called discriminant analysis it is possible to determine on the basis of any set of STAR sequences in a species, the most optimal discriminative sequence patterns or combination thereof. Thus, preferably, at least one of the patterns is selected on the basis of optimal discrimination between at least one sequence comprising a STAR sequence and a control sequence.
In a preferred embodiment, the frequency of occurrence of a sequence pattern in a test nucleic acid is compared with the frequency of occurrence in a sequence known to contain a STAR sequence. In this case, a pattern is considered representative for a sequence comprising a STAR sequence if the frequencies of occurrence are similar. In a preferred embodiment, another criterion is used. The frequency of occurrence of a pattern in a sequence comprising a STAR sequence is compared to the frequency of occurrence of the pattern in a control sequence. By comparing the two frequencies it is possible to determine for each pattern thus analyzed, whether the frequency in the sequence comprising the STAR sequence is significantly different from the frequency in the control sequence. In this embodiment, a sequence pattern is considered to be representative of a sequence comprising a STAR sequence, if the frequency of occurrence of the pattern in at least one sequence comprising a STAR sequence is significantly different from the frequency of occurrence of the same pattern in a control sequence. By using larger numbers of sequences comprising a STAR sequence the number of patterns for which a statistical difference can be established increases, thus enlarging the number of patterns for which the frequency of occurrence is representative for a sequence comprising a STAR sequence. Preferably, the frequency of occurrence is representative of the frequency of occurrence of at least one sequence pattern in at least two sequences comprising a STAR sequence; more preferably, in at least five sequences comprising a STAR sequence; and, even more preferably, in at least ten sequences comprising a STAR sequence. More preferably, the frequency of occurrence is representative of the frequency of occurrence of at least one sequence pattern in at least 20 sequences comprising a STAR sequence. In a particularly preferred embodiment, the frequency of occurrence is representative of the frequency of occurrence of at least one sequence pattern in at least 50 sequences comprising a STAR.
The patterns that are indicative for a sequence comprising a STAR sequence are also dependent on the type of control nucleic acid used. The type of control sequence used is preferably selected on the basis of the sequence in which the presence of a STAR sequence is to be detected. In a preferred embodiment, the control sequence comprises a random sequence comprising a similar AT/CG content as at least one sequence comprising a STAR sequence. In another preferred embodiment, the control sequence is derived from the same species as the sequence comprising the STAR sequence. For instance, if a test sequence is scrutinized for the presence of a STAR sequence, active in a plant cell, then preferably the control sequence is also derived from a plant cell. Similarly, for testing for STAR activity in a human cell, the control nucleic acid is preferably also derived from a human genome. In a preferred embodiment, the control sequence comprises between 50% and 150% of the bases of at least one sequence comprising a STAR sequence. In a particularly preferred embodiment, the control sequence comprises between 90% and 110% of the bases of at least one sequence comprising a STAR sequence. More preferably, between 95% and 105%.
A pattern can comprise any number of bases larger than two. Preferably, at least one sequence pattern comprises at least five, more preferably at least six, bases. In another embodiment, at least one sequence pattern comprises at least eight bases. In a preferred embodiment, the at least one sequence pattern comprises a pattern listed in Table 6 and/or Table 7. A pattern may consist of a consecutive list of bases. However, the pattern may also comprise bases that are interrupted one or more times by a number of bases that are not or only partly discriminative. A partly discriminative base is, for instance, indicated as a purine.
Preferably, the presence of STAR activity is verified using a functional assay. Several methods are presented herein to determine whether a sequence comprises STAR activity. STAR activity is confirmed if the sequence is capable of performing at least one of the following functions: (i) at least in part inhibiting the effect of sequence comprising a gene transcription repressing element of the invention, (ii) at least in part blocking chromatin-associated repression, (iii) at least in part blocking activity of an enhancer, (iv) conferring upon an operably linked nucleic acid encoding a transcription unit compared to the same nucleic acid alone, (iv-a) a higher predictability of transcription, (iv-b) a higher transcription, and/or (iv-c) a higher stability of transcription over time.
The large number of sequences comprising STAR activity identified by the present inventors open up a wide variety of possibilities to generate and identify sequences comprising the same activity in kind not necessarily in amount. For instance, it is well within the reach of a skilled person to alter the sequences identified in the present invention and test the altered sequence for STAR activity. Such altered sequences are, therefore, also part of the present invention. Alteration can include deletion, insertion and mutation of one or more bases in the sequences.
Sequences comprising STAR activity were identified in stretches of 400 bases. However, it is expected that not all of these 400 bases are required to retain STAR activity. Methods to delimit the sequences that confer a certain property to a fragment of between 400 and 5000 bases are well known. The minimal sequence length of a fragment comprising STAR activity is estimated to be about 50 bases.
Table 6 (SEQ ID NOS:177-342) and Table 7 (SEQ ID NOS:343-1072) list patterns of six bases that have been found to be over represented in nucleic acid molecules comprising STAR activity. This over representation is considered to be representative for a STAR sequence. The tables were generated for a family of 65 STAR sequences (SEQ ID NOS:1-65). Similar tables can be generated starting from a different set of STAR sequences, or from a smaller or larger set of STAR sequences. A pattern is representative for a STAR sequence if it is over represented in the STAR sequence compared to a sequence not comprising a STAR element. This can be a random sequence. However, to exclude a non relevant bias, the sequence comprising a STAR sequence is preferably compared to a genome or a significant part thereof. Preferably, a genome of a vertebrate or plant, more preferably, a human genome. A significant part of a genome is, for instance, a chromosome. Preferably the sequence comprising a STAR sequence and the control sequence are derived from nucleic acid of the same species.
The more STAR sequences are used for the determination of the frequency of occurrence of sequence patterns, the more representative for STARs the patterns are that are over- or under-represented. Considering that many of the functional features that can be expressed by nucleic acids are mediated by proteinaceous molecules binding to them, it is preferred that the representative pattern is over-represented in the STAR sequences. Such over-represented pattern can be part of a binding site for such a proteinaceous molecule. Preferably, the frequency of occurrence is representative of the frequency of occurrence of at least one sequence pattern in at least two sequences comprising a STAR sequence; more preferably, in at least five sequences comprising a STAR sequence; and, even more preferably, in at least ten sequences comprising a STAR sequence. More preferably, the frequency of occurrence is representative of the frequency of occurrence of at least one sequence pattern in at least 20 sequences comprising a STAR sequence. In a particularly preferred embodiment, the frequency of occurrence is representative of the frequency of occurrence of at least one sequence pattern in at least 50 sequences comprising a STAR. Preferably, the sequences comprising a STAR sequence comprises at least one of the sequences depicted in the sequences comprising STAR1-STAR65 (SEQ ID NOS:1-65), sequences comprising STAR66 and testing set (SEQ ID NOS:66-84), and sequences comprising Arabidopsis STAR A1-A35 (SEQ ID NOS:85-119) (hereinafter SEQ ID NOS:1-119).
STAR activity is a feature shared by the sequences listed in SEQ ID NOS:1-119. However, this does not mean that they must all share the same identifier sequence. It is very well possible that different identifiers exist. Identifiers may confer this common feature onto a fragment containing it, though this is not necessarily so.
By using more sequences comprising STAR activity for determining the frequency of occurrence of a sequence pattern or patterns, it is possible to select patterns that are more often than others present or absent in such a STAR sequence. In this way it is possible to find patterns that are very frequently over- or under-represented in STAR sequences. Frequently, over- or under-represented patterns are more likely to identify candidate STAR sequences in test sets. Another way of using a set of over- or under-represented patterns is to determine which pattern or combination of patterns is best suited to identify a STAR in a sequence. Using so-called discriminative statistics, we have identified a set of patterns that performs best in identifying a sequence comprising a STAR element. In a preferred embodiment, at least one of the sequence patterns for detecting a STAR sequence comprises a sequence pattern GGACCC (SEQ ID NO:464), CCCTGC (SEQ ID NO:816), AAGCCC (SEQ ID NO:270), CCCCCA (SEQ ID NO:298) and/or AGCACC (SEQ ID NO:336). In another embodiment, at least one of the sequence patterns for detecting a STAR sequence comprises a sequence pattern CCCN{16}AGC (SEQ ID NO:415), GGCN{9}GAC (SEQ ID NO:536), CACN{13}AGG (SEQ ID NO:761), and/or CTGN{4}GCC (SEQ ID NO:839).
A list of STAR sequences can also be used to determine one or more consensus sequences therein. The invention, therefore, also provides a consensus sequence for a STAR element. This consensus sequence can of course be used to identify candidate STAR elements in a test sequence.
Moreover, once a sequence comprising a STAR element has been identified in a vertebrate it can be used by means of sequence homology to identify sequences comprising a STAR element in other species belonging to vertebrate. Preferably a mammalian STAR sequence is used to screen for STAR sequences in other mammalian species. Similarly, once a STAR sequence has been identified in a plant species, it can be used to screen for homologous sequences with similar function in other plant species. The invention in one aspect provides a STAR sequence obtainable by a method according to the invention. Further provided is a collection of STAR sequences. Preferably, the STAR sequence is a vertebrate or plant STAR sequence. More preferably, the STAR sequence is a mammalian STAR sequence or an angiosperm (monocot, such as rice or dicot, such as Arabidopsis). More preferably, the STAR sequence is a primate and/or human STAR sequence.
A list of sequences comprising STAR activity can be used to determine whether a test sequence comprises a STAR element. There are, as mentioned above, many different methods for using such a list for this purpose. In a preferred embodiment, the invention provides a method for determining whether a nucleic acid sequence of about 50-5000 base pairs comprises a STAR sequence, the method comprising: generating a first table of sequence patterns comprising the frequency of occurrence of the patterns in a collection of STAR sequences of the invention; generating a second table of the patterns comprising the frequency of occurrence of the patterns in at least one reference sequence; selecting at least one pattern of which the frequency of occurrence differs between the two tables; determining, within the nucleic acid sequence of about 50-5000 base pairs, the frequency of occurrence of at least one of the selected patterns; and determining whether the occurrence in the test nucleic acid is representative of the occurrence of the selected pattern in the collection of STAR sequences. Alternatively, determining comprises determining whether the frequency of occurrence in the test nucleic acid is representative of the frequency occurrence of the selected pattern in the collection of STAR sequences. Preferably, the method further comprises determining whether the candidate STAR comprises a gene transcription modulating quality using a method of the invention. Preferably, the collection of STARS comprises sequence as depicted in SEQ ID NOS:1-119. In another aspect, the invention provides an isolated and/or recombinant nucleic acid sequence comprising a STAR sequence obtainable by a method of the invention.
As mentioned above, a STAR sequence can exert its activity in a directional way, i.e., more to one side of the fragment containing it than to the other. Moreover, STAR activity can be amplified in amount by multiplying the number of STAR elements. The latter suggests that a STAR element may comprise one or more elements comprising STAR activity. Another way of identifying a sequence capable of conferring STAR activity on a fragment containing it comprises selecting from a vertebrate or plant sequence, a sequence comprising STAR activity and identifying whether the selected sequence and sequences flanking the selected sequence are conserved in another species. Such conserved flanking sequences are likely to be functional sequences. In one aspect, the invention, therefore, provides a method for identifying a sequence comprising a STAR element comprising selecting a sequence of about 50 to 5000 base pairs from a vertebrate or plant species comprising a STAR element and identifying whether sequences flanking the selected sequence in the species are conserved in at least one other species. The invention, therefore, further provides a method for detecting the presence of a STAR sequence within a nucleic acid sequence of about 50-5000 base pairs, comprising identifying a sequence comprising a STAR sequence in a part of a chromosome of a cell of a species and detecting significant homology between the sequence and a sequence of a chromosome of a different species. Preferably, the species comprises a plant or vertebrate species, ideally a mammalian species. The invention also provides a method for detecting the presence of a STAR element within a nucleic acid sequence of about 50-5000 base pairs of a vertebrate or plant species, comprising identifying whether a flanking sequence of the nucleic acid sequence is conserved in at least one other species.
It is important to note that methods of the invention for detecting the presence of a sequence comprising a STAR sequence using bioinformatical information are iterative in nature. The more sequences comprising a STAR sequence are identified with a method of the invention, the more patterns are found to be discriminative between a sequence comprising a STAR sequence and a control sequence. Using these newly found discriminative patterns, more sequences comprising a STAR sequence can be identified, which, in turn, enlarges the set of patterns that can discriminate and so on. This iterative aspect is an important aspect of methods provided in the present invention.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 is a diagram illustrating the pSelect plasmid used for isolating STAR elements. The zeocin resistance gene is under control of the SV40 promoter, and is upstream of the SV40 polyadenylation site. Upstream of the SV40 promoter is a tandem array of lexA operator sites. Between the lexA operators and the SV40 promoter is a cloning site; test DNAs (e.g., size-fractionated genomic DNA) are cloned into the BamHI site. The plasmid also has the hygromycin resistance gene (hygro) for selection of transformed cells, the EBNA-1 and oriP sequences for plasmid replication in mammalian cells, and the ampicillin resistance gene (ampR) and ori sequence for propagation in Escherichia coli.
FIG. 2 is a diagram depicting the pSDH-CSP plasmid used for testing STAR activity. The Secreted Alkaline Phosphatase (SEAP) reporter gene is under control of the CMV promoter, and the puromycin resistance selectable marker (puro) is under control of the SV40 promoter. Flanking these two genes are multiple cloning sites (MCSI and MCSII) into which STAR elements can be cloned. The plasmid also has an origin of replication (ori) and ampicillin resistance gene (ampR) for propagation in Escherichia coli.
FIG. 3 is a graph showing that STAR6 (SEQ ID NO:6) and STAR8 (SEQ ID NO:8) improve predictability and yield of transgene expression in U-2 OS cells. Expression of luciferase from the CMV promoter by U-2 OS cells transfected with pSDH-CMV, pSDH-CMV-STAR6, or pSDH-CMV-STAR8 was determined. The STAR-containing constructs confer greater predictability and elevated yield relative to the pSDH-CMV construct alone.
FIG. 4 is a graph illustrating that STAR6 (SEQ ID NO:6), but not STAR8 (SEQ ID NO:8), improves predictability and yield of transgene expression in CHO cells. Expression of SEAP from the CMV promoter by CHO cells transfected with pSDH-CSP, pSDH-CSP-STAR6, or pSDH-CSP-STAR8 was determined. The STAR6-containing constructs confer greater predictability and elevated yield relative to the pSDH-CSP construct alone, identifying STAR6 (SEQ ID NO:6) as a promiscuous STAR element. In contrast, the STAR8-containing constructs do not consistently increase yield or predictability relative to the pSDH-CSP construct, suggesting that STAR8 (SEQ ID NO:8) is a cell line-specific STAR element.
FIG. 5 is a graph depicting that STAR6 (SEQ ID NO:6) and STAR49 (SEQ ID NO:49) improve predictability and yield of transgene expression. Expression of SEAP from the CMV promoter by CHO cells transfected with pSDH-CSP, pSDH-CSP-STAR6, or pSDH-CSP-STAR49 was determined. The STAR-containing constructs confer greater predictability and elevated yield relative to the pSDH-CSP construct alone.
FIG. 6 is a graph showing the minimal essential sequences of STAR10 (SEQ ID NO:10) and STAR27 (SEQ ID NO:27). Portions of the STAR elements were amplified by PCR: STAR10 (SEQ ID NO:10) was amplified with primers E23 (SEQ ID NO:166) and E12 (SEQ ID NO:155) to yield fragment 10A (corresponding approximately to the first 400 nucleotides of SEQ ID NO:10), E13 (SEQ ID NO:156) and E14 (SEQ ID NO:157) to yield fragment 10B (corresponding approximately to the second 400 nucleotides of SEQ ID NO:10), and E15 (SEQ ID NO:158) and E16 (SEQ ID NO:159) to yield fragment 10C (corresponding approximately to the third 400 nucleotides of SEQ ID NO:10). STAR27 (SEQ ID NO:27) was amplified with primers E17 (SEQ ID NO:160) and E18 (SEQ ID NO:161) to yield fragment 27A (corresponding approximately to the first 500 nucleotides of SEQ ID NO:27), E19 (SEQ ID NO:162) and E20 (SEQ ID NO:163) to yield fragment 27B (corresponding to the second 500 nucleotides of SEQ ID NO:27), and E21 (SEQ ID NO:164) and E22 (SEQ ID NO:165) to yield fragment 27C (corresponding approximately to the third 500 nucleotides of SEQ ID NO:27). These sub-fragments were cloned into the pSelect vector. After transfection into U-2 OS/Tet-Off/LexA-HP1 cells, the growth of the cultures in the presence of zeocin was monitored. Growth rates varied from vigorous (+++) to poor (+/−), while some cultures failed to survive zeocin treatment (−) due to absence of STAR activity in the DNA fragment tested.
FIG. 7 is a graph illustrating the STAR element function in the context of the SV40 promoter. pSDH-SV40 and pSDH-SV40-STAR6 were transfected into the human osteosarcoma U-2 OS cell line, and expression of luciferase was assayed with or without protection from gene silencing by STAR6 (SEQ ID NO:6) in puromycin-resistant clones.
FIG. 8 is a graph showing the STAR element function in the context of the Tet-Off promoter. pSDH-Tet and pSDH-Tet-STAR6 were transfected into the human osteosarcoma U-2 OS cell line, and expression of luciferase was assayed with or without protection from gene silencing by STAR6 (SEQ ID NO:6) in puromycin-resistant clones.
FIG. 9 is a schematic diagram of the orientation of STAR elements as they are cloned in the pSelect vector (panel A), as they are cloned into pSDH vectors to preserve their native orientation (panel B), and as they are cloned into pSDH vector in the opposite orientation (panel C).
FIG. 10 is a graph showing directionality of STAR66 (SEQ ID NO:66) function. The STAR66 (SEQ ID NO:66) element was cloned into pSDH-Tet in either the native (STAR66 native) or the opposite orientation (STAR66 opposite) and transfected into U-2 OS cells. Luciferase activity was assayed in puromycin resistant clones.
FIG. 11 is a southern blot showing copy number-dependence of STAR function. Southern blot of luciferase expression units in pSDH-Tet-STAR10, integrated into U-2 OS genomic DNA. Radioactive luciferase DNA probe was used to detect the amount of transgene DNA in the genome of each clone, which was then quantified with a phosphorimager.
FIG. 12 is a graph illustrating copy number-dependence of STAR function. The copy number of pSDH-Tet-STAR10 expression units in each clone was determined by phosphorimagery and compared with the activity of the luciferase reporter enzyme expressed by each clone.
FIG. 13 is a schematic diagram and graphs depicting enhancer-blocking and enhancer assays. The luciferase expression vectors used for testing STARs for enhancer-blocking and enhancer activity are shown schematically. The E-box binding site for the E47 enhancer protein is upstream of a cloning site for STAR elements. Downstream of the STAR cloning site is the luciferase gene under control of a human alkaline phosphatase minimal promoter (mp). The histograms indicate the expected outcomes for the three possible experimental situations (see text). Panel A: Enhancer-blocking assay. Panel B: Enhancer assay.
FIG. 14 is a graph showing enhancer-blocking assay. Luciferase expression from a minimal promoter is activated by the E47/E-box enhancer in the empty vector (vector). Insertion of enhancer-blockers (scs, HS4) or STAR elements ( STAR elements 1, 2, 3, 6, 10, 11, 18, and 27; SEQ ID NOS:1, 2, 3, 6, 10, 11, 18 and 27, respectively) block luciferase activation by the E47/E-box enhancer.
FIG. 15 is a graph illustrating enhancer assay. Luciferase expression from a minimal promoter is activated by the E47/E-box enhancer in the empty vector (E47). Insertion of the scs and HS4 elements or various STAR elements ( STARs 1, 2, 3, 6, 10, 11, 18, and 27; SEQ ID NOS:1, 2, 3, 6, 10, 11, 18 and 27, respectively) do not activate transcription of the reporter gene.
FIG. 16 illustrates STAR18 (SEQ ID NO:18) sequence conservation between mouse and human. The region of the human genome containing 497 base pair STAR18 (SEQ ID NO:18) is shown (black boxes); the element occurs between the HOXD8 and HOXD4 homeobox genes on human chromosome 2. It is aligned with a region in mouse chromosome 2 that shares 72% sequence identity. The region of human chromosome 2 immediately to the left of STAR18 (SEQ ID NO:18) is also highly conserved with mouse chromosome 2 (73% identity; gray boxes); beyond these region, the identity drops below 60%. The ability of these regions from human and mouse, either separately or in combination, to confer growth on zeocin is indicated: −, no growth; +, moderate growth; ++, vigorous growth; +++, rapid growth.
FIG. 17 is a schematic diagram of bio-informatic analysis workflow. For details, see text.
FIG. 18 is a schematic diagram showing the results of discriminant analysis on classification of the training set of 65 STAR elements. STAR elements that are correctly classified as STARs by Stepwise Linear Discriminant Analysis (LDA) are shown in a Venn diagram. The variables for LDA were selected from frequency analysis results for hexameric oligonucleotides (“oligos”) and for dyads. The diagram indicates the concordance of the two sets of variables in correctly classifying STARs.
FIG. 19 is a graph illustrating that U-2 OS/Tet-Off/lexA-HP1 cells were transfected with candidate Arabidopsis STAR elements and cultivated at low doxycycline concentrations. Total RNA was isolated and subjected to RT-PCR; the bands corresponding to the zeocin and hygromycin resistance mRNAs were detected by Southern blotting and quantified with a phosphorimager. The ratio of the zeocin to hygromycin signals is shown for transfectants containing zeocin expression units flanked by 12 different Arabidopsis STAR elements, the Drosophila scs element, or no flanking element.
FIG. 20 is a schematic diagram and graph illustrating that STAR elements improve GFP expression in CHO cells. The ppGIZ and ppGIZ-STAR7 plasmids used for testing STAR activity are shown. The expression unit comprises (from 5′ to 3′) a transgene (encoding for the GFP protein), an IRES, and a selectable marker (zeo, conferring zeocin resistance) under control of the CMV promoter. The expression unit has the SV40 transcriptional terminator at its 3′ end (t). The entire cassette with the expression unit is either flanked by STAR7 (SEQ ID NO:7) elements (STAR7-shielded) or not (Control). The constructs are transfected to CHO-K1 cells. Stable colonies are expanded and the GFP signal is determined on a XL-MCL Beckman Coulter flow cytometer. For each independent colony the mean of the GFP signal is plotted. This is taken as measure for the level of GFP expression. The results in FIG. 20 show that in CHO cells the STAR7-shielded construct confers greater predictability and elevated GFP expression relative to the ppGIZ control construct alone.
FIG. 21 is a schematic diagram and graph showing that STAR elements improve GFP expression in NSO cells. The ppGIZ and ppGIZ-STAR7 plasmids used for testing STAR activity are shown as in FIG. 20. The constructs are transfected to NSO cells. Stable colonies are expanded and the GFP signal is determined on a XL-MCL Beckman Coulter flow cytometer. For each independent colony the mean of the GFP signal is plotted. This is taken as measure for the level of GFP expression. The results in FIG. 21 show that in NSO cells the STAR7-shielded (SEQ ID NO:7) construct confers greater predictability and elevated GFP expression relative to the ppGIZ control construct alone.
FIG. 22 is a schematic diagram and graph depicting that STAR elements improve GFP expression in 293 cells. The ppGIZ and ppGIZ-STAR7 plasmids used for testing STAR activity are shown as in FIG. 20. The constructs are transfected to 293 cells. Stable colonies are expanded and the GFP signal is determined on a XL-MCL Beckman Coulter flow cytometer. For each independent colony, the mean of the GFP signal is plotted. This is taken as measure for the level of GFP expression. The results in FIG. 22 show that in 293 cells the STAR7-shielded (SEQ ID NO:7) construct confers greater predictability and elevated GFP expression relative to the ppGIZ control construct alone.
DETAILED DESCRIPTION OF THE INVENTION Definitions
Quality
The term “quality” in relation to a sequence refers to an activity of the sequence.
STAR and STAR Sequence
The terms “STAR,” “STAR sequence” or “STAR element,” as used herein, refer to a DNA sequence comprising one or more of the mentioned gene transcription modulating qualities.
DNA Sequence
The term “DNA sequence” as used herein, unless otherwise specified, does not refer to a listing of specific ordering of bases, but rather to a physical piece of DNA. A transcription quality with reference to a DNA sequence refers to an effect that the DNA sequence has on transcription of a gene of interest. “Quality” as used herein refers to detectable properties or attributes of a nucleic acid or protein in a transcription system.
Proteinaceous Molecule
By a “proteinaceous molecule” is meant herein a molecule comprising amino acids. At least a part of the amino acids are bound to each other to form a peptide. Preferably, the proteinaceous molecule comprises a polypeptide. In this application, the term “proteinaceous molecule” also includes “polypeptide.”
Essentially the Same Properties
By “essentially the same properties” is meant that the properties are essentially the same in kind, not necessarily in amount. For instance, if a proteinaceous molecule has essentially the same properties as a pharmaceutically active compound, the proteinaceous molecule also displays such pharmaceutical activity in kind, not necessarily in amount.
Naturally Occurring Proteinaceous Molecule of the Same Kind
By a “naturally occurring proteinaceous molecule of the same kind” is meant a proteinaceous molecule with the same primary structure, which is naturally produced in vivo, not influenced by human interference. Examples comprise an antibody produced in vivo by a lymphocyte and erythropoietin produced in vivo by a hepatocyte.
Host Cell, Host Cell Line
As used herein, the terms “host cell” and “host cell line” refer to a cell and to homogeneous populations thereof that are capable of expressing a nucleic acid encoding a proteinaceous molecule.
Recombinant Host Cell, Recombinant Host Cell Line
The terms “recombinant host cell” and “recombinant host cell line” refer to a host cell and to homogeneous populations thereof into which a nucleic acid has been introduced.
Expression
As used herein, the term “expression” refers to production of a proteinaceous molecule, encoded by a nucleic acid. The production, for instance, involves transcription of a DNA sequence, translation of the corresponding mRNA sequence, and/or posttranslational modification. In case of secreted proteins, it can also refer to the processes of transcription, translation, and/or post-translational modification (e.g., glycosylation, disulfide bond formation, etc.), followed by exocytosis. In the case of multimeric proteins, it can include assembly of the multimeric structure from the polypeptide monomers.
Silencing
The term “silencing” refers to diminution of a level of expression of a gene or genes, including transgenes, typically over time. The expression level can be diminished but still detectable, or diminished below the threshold of detection.
Enhanced Expression
As used herein, “enhanced expression” of a gene encoding a proteinaceous molecule, or enhanced production of a proteinaceous molecule, can either involve a higher yield of the proteinaceous molecule, a higher proportion of host cells with acceptable expression levels, and/or a higher stability of a gene expression level.
Affiliated Proteinaceous Molecule of a Cell
By an “affiliated proteinaceous molecule of a cell” is meant a proteinaceous molecule which is naturally produced by the kind of cell in the organism from which the cell is derived.
For instance, erythropoietin is an affiliated proteinaceous molecule of a hepatocyte, or of a hepatocyte-derived cell line. Likewise, an antibody is an affiliated proteinaceous molecule of a lymphocyte, or of a lymphocyte-derived cell line, typically of a B-cell or a B-cell derived cell line.
Specific Host Cell Line
A “specific host cell line” is a host cell line derived from a cell which normally expresses a particular proteinaceous molecule (or class of proteinaceous molecules) in the organism from which the cell is derived.
Heterologous STAR Sequence
The term “heterologous STAR sequence” is herein used to define a STAR sequence that is, for example, obtained from a different cell type (from the same species or organism) or is obtained from a different species or organism (either from the same cell type or a different cell type) compared to the cell in which it is used.
Stable
“Stable” means that the observed transcription level is not significantly changed over at least 30 cell divisions. A stable quality is useful in situations wherein expression characteristics should be predictable over many cell divisions. Typical examples are cell lines transfected with foreign genes. Other examples are transgenic animals and plants and gene therapies. Very often, introduced expression cassettes function differently after increasing numbers of cell divisions or plant or animal generations. A stable quality preferably comprises a capacity to maintain gene transcription in subsequent generations of a transgenic plant or animal. Of course, in the case where expression is inducible, the quality comprises the quality to maintain inducibility of expression in subsequent generations of a transgenic plant or animal.
Acceptable Expression Level
An “acceptable expression level” means an acceptable expression level for commercial exploitation. Whether or not a certain expression level is acceptable for commercial exploitation often depends on the kind of proteinaceous molecule that is produced. Acceptable expression levels of different kinds of proteinaceous molecules often involve different amounts of produced proteinaceous molecule.
A STAR sequence, a collection of STAR sequences, and/or a nucleic acid comprising a STAR sequence obtainable by a method of the invention, is of course suitable for use in a method of the invention for producing a proteinaceous molecule in a cell. In one aspect, the invention, therefore, provides a method for producing a proteinaceous molecule in a cell comprising selecting a cell for its suitability for producing the proteinaceous molecule, providing a nucleic acid encoding the proteinaceous molecule with a nucleic acid comprising a STAR sequence, expressing the resulting nucleic acid in the cell and collecting the proteinaceous molecule, wherein the nucleic acid comprising a STAR sequence is obtainable by a method of the invention for identifying and obtaining a nucleic acid comprising a STAR sequence. The nucleic acid comprising a STAR sequence can be identified and obtained using at least one pattern that is representative for sequences comprising STAR activity. Preferably, the nucleic acid comprising a STAR sequence is identified and obtained using at least one pattern as depicted in Table 6 (SEQ ID NOS:177-342) or Table 7 (SEQ ID NOS:343-1072).
A cell can be selected for its suitability for producing a proteinaceous molecule in many different ways. For instance, it can be determined whether the cell is competent of nucleic acid uptake. A nucleic acid encoding a proteinaceous molecule is preferably easily introduced into the cell. Furthermore it can be determined whether the cell secretes produced proteinaceous molecule. Secreted proteinaceous molecule can usually be easily collected. Collecting proteinaceous molecules that are not secreted usually involves sacrificing at least part of a culture. This implicates separating a proteinaceous molecule of interest from other cell components, optionally starting up a new culture, etc. This is more cumbersome. Hence, in a preferred embodiment, a method of the invention is provided wherein the proteinaceous molecule is secreted by the cell.
In another preferred embodiment, a method of the invention is provided wherein the cell is selected for its capability of post-translationally modifying the proteinaceous molecule, such that the proteinaceous molecule has essentially the same properties as a naturally occurring proteinaceous molecule of the same kind. As has been explained above, different kinds of cells display different post-translational modifications. As a result, the same proteinaceous molecule can be modified differently when produced in different host cells. These differences can affect the properties of such proteinaceous molecules, such as for instance pharmaceutical properties. It is, therefore, highly preferred to select a cell which produces a proteinaceous molecule with essentially the same properties as its naturally occurring counterpart. This does not necessarily mean that the host cell has to display exactly the same kind of post-translational modifications, as long as the produced proteinaceous molecule has essentially the same properties. A proteinaceous molecule can be produced that is physically different from its natural counterpart, but which is functionally essentially the same.
In one embodiment, of the invention, however, a cell is selected for its capability of post-translationally modifying the proteinaceous molecule in essentially the same way as the proteinaceous molecule is post-translationally modified in nature.
In one aspect, the invention provides a method of the invention wherein the proteinaceous molecule comprises an affiliated proteinaceous molecule of the cell.
It is especially preferred to choose a host cell, or a cell line derived from a cell, which normally produces a proteinaceous molecule of interest in the organism from which the cell is derived. These cells are capable of carrying out post-translational modifications of their affiliated proteinaceous molecules such that the resulting proteinaceous molecule has the same kind of properties in kind, not necessarily in amount, as a proteinaceous molecule of the same kind which is normally present in the organism. Such specific cells are naturally adapted for production of the affiliated proteinaceous molecule. Apart from other activities of STAR sequences it is also possible to at least partly solve a problem of low production of proteinaceous molecules, which often occurs with such specific cells. Providing at least one STAR sequence to a nucleic acid encoding such proteinaceous molecule will enhance production (yield) of the proteinaceous molecule by the specific cell, increase the proportion of host cells with acceptable expression levels, and/or increase stability of a gene expression level.
In another preferred embodiment, a method of the invention is disclosed wherein the cell is selected for suitable growth in a suspension culture. This facilitates culturing of the cell, and collection of produced proteinaceous molecule, especially when the proteinaceous molecule is secreted into the suspension. More preferably, the cell is selected for suitable growth in a serum-free culture, since serum can contain contaminants and pathogens. Such contaminants/pathogens often have to be separated from produced proteinaceous molecule. This requires an extra step, which consumes time and chemicals, with a potential loss of produced proteinaceous molecule. Moreover, a possibility of a presence of pathogens involves a potential risk for employees. If a pathogen has been present in a culture, a produced proteinaceous molecule is not allowed to be used anymore for commercial and/or medical applications.
In yet another preferred embodiment, the cell is selected for the presence of an adenovirus Early Region 1 (E1) sequence. The presence of an adenoviral E1 sequence enhances protein production in a cell. Hence, an adenoviral E1 sequence is suitable for host cells engineered for protein production.
The invention also provides a method for producing a proteinaceous molecule comprising:
    • providing a host cell with a nucleic acid comprising a STAR sequence;
    • selecting a cell with enhanced expression of a proteinaceous molecule; and
    • collecting the proteinaceous molecule.
In one embodiment, the method is performed with a host cell whose genome has not been modified by human interference. The method then results in enhanced expression of a proteinaceous molecule which is encoded by the genome of the host cell. The proteinaceous molecule may be normally expressed by the host cell in the organism from which the cell is derived, but it may also normally be subject to silencing, resulting in little or no expression in the host cell under normal conditions. Introduction of at least one STAR sequence can at least in part inhibit silencing of a gene of interest induced by gene-transcription repressing chromatin. Expression of a proteinaceous molecule is enhanced by introduction of a STAR sequence.
In another embodiment, the host cell is transfected with a nucleic acid of interest. Such nucleic acid for instance, encodes a heterologous proteinaceous molecule which is not naturally encoded by the genome of the host cell. Introduction of a STAR sequence also enhances expression of such heterologous proteinaceous molecule.
The STAR sequence can be introduced randomly into the genome of the host cell, using methods known in the art (for instance calcium precipitation, transfection with a vector comprising a nucleic acid of interest, use of a gene delivery vehicle, etc). If a STAR sequence is introduced near a nucleic acid sequence encoding a proteinaceous molecule, it is capable of enhancing expression of the proteinaceous molecule. Cells expressing a desired proteinaceous molecule can be isolated from cultures with randomly inserted STAR sequences.
Preferably, the STAR sequence is introduced into the host cell by homologous recombination. A nucleic acid comprising a STAR sequence can be provided with an additional sequence. The additional sequence can be chosen such that it is at least in part homologous to a nucleic acid sequence in the host cell which is known to be present in vicinity of a gene encoding a proteinaceous molecule of interest. If a nucleic acid comprising a STAR sequence and such additional sequence is provided to the host cell, it can be incorporated into the host cell's genome by homologous recombination at the site with the (partly) homologous nucleic acid sequence. As a result, the STAR sequence is introduced in vicinity of the gene encoding the proteinaceous molecule of interest. Expression of the proteinaceous molecule is then enhanced by the introduced STAR sequence.
A preferred embodiment of the present invention provides a method of the invention wherein the STAR sequence comprises a species-specific STAR sequence. More preferably, the STAR sequence comprises a cell type-specific STAR sequence.
Two types of STAR elements have been identified. Promiscuous STAR elements are able to function in more than one host cell line. For example, STAR6 (SEQ ID NO:6) increases the predictability, yield, and stability of a transgene in both the U-2 OS human osteosarcoma cell line and in CHO (Chinese hamster ovary) cells. Other STAR elements are species-specific and/or cell type-specific; for example, STAR8 (SEQ ID NO:8) increases the predictability, yield, and stability of transgenes in U-2 OS cells, but not in CHO cells (see Examples 2 and 3 and FIGS. 3 and 4).
If a certain type of host cell (line) is chosen for expression of a proteinaceous molecule (for instance, because it is known to possess a preferred post-translational modification system) a STAR sequence which is naturally present in the cell can be used in a method of the invention. Such STAR sequence is referred to as a cell type-specific STAR sequence. A STAR sequence which is naturally present in a species from which the cell is derived can also be used. Such STAR sequence is referred to as a species-specific STAR sequence. A species-specific STAR sequence may be naturally present in the cell type, although this is not necessary.
A known species-specific STAR sequence or cell-type specific STAR sequence can be used in a method of the invention. Alternatively, a (previously unknown) species-specific STAR sequence or cell-type specific STAR sequence can be detected and isolated by a method as described by the present inventors (EP 01202581.3). The use of a species-specific STAR sequence or cell type-specific STAR sequence is preferred because such sequence is especially active in the host cell and is adapted to the specific circumstances within the cell. For instance, such cell type-specific STAR sequence may interact with a protein which is not present in some other cell-types. In that case, the cell type-specific STAR sequence will be less capable—if at all—of enhancing expression in cells lacking the protein. A species-specific or cell type-specific STAR element often has functional characteristics that are superior to promiscuous STAR elements. Furthermore, a cell line-specific STAR element can satisfy product safety or ethical considerations for use of the host cell line.
A promiscuous STAR sequence is particularly useful if no tissue specific or cell-type specific STAR sequence is known. In that case a known promiscuous STAR sequence can be used. This saves efforts to detect and isolate a cell-type specific STAR sequence.
Several STAR sequences are listed in SEQ ID NOS:1-119. Hence, in one aspect, a method of the invention is provided wherein the STAR sequence comprises a sequence as depicted in SEQ ID NOS:1-119.
In a preferred embodiment, the invention provides a cell line that comprises at least one heterologous STAR sequence or a functional equivalent and/or a functional fragment thereof. In an even more preferred embodiment, the cell line is a human cell line. The invention provides multiple examples of STAR sequences and also methods of testing STAR sequences and hence, a person skilled in the art is very well capable of obtaining a functional equivalent and/or a functional fragment of a STAR sequence, for example by deletion or mutation. In yet another preferred embodiment, the invention provides a non-human cell line that comprises at least one recombinant STAR sequence derived or obtained from a human cell, i.e., a human STAR sequence. It is clear that the amount of STAR sequences may vary, for example, a cell line according to the invention may comprise two, three, or four, or even more STAR sequences which may either be identical or different from each other.
In one aspect, the invention provides a cell line provided with a nucleic acid comprising a STAR sequence, wherein the cell line is selected for its suitability for producing a proteinaceous molecule. Preferably, a cell line of the invention comprises a vertebrate or plant cell line. A vertebrate cell line is very suitable for producing a human proteinaceous molecule of interest, because vertebrates are phylogenetically close related.
Plant cells are for instance very suitable for vaccine production. Vaccine production in plants can be inexpensive, while the vaccine can be easily delivered to an individual by eating the edible portion of the plant (Mercenier et al., 2001).
A cell line of the invention is particularly suitable for production of a proteinaceous molecule of interest, because the STAR sequence can enhance expression of a gene of interest (higher yield of a proteinaceous molecule, higher proportion of host cells with acceptable expression levels, and/or higher stability of a gene expression level). Methods for generating a cell line are known in the art and many techniques are known to provide a cell with a nucleic acid of interest. Furthermore, many general purpose cell lines are available. Such cell lines can be dedicated to production of a certain proteinaceous molecule using recombinant techniques. Examples of available cell lines include CHO cells from Chinese hamster ovary and BHK cells from baby hamster kidney (as described above).
Another embodiment of the invention provides a cell line provided with a nucleic acid comprising a STAR sequence, wherein the cell line comprises an adenovirus Early Region 1 sequence. As has been described above, an adenoviral E1 sequence enhances cellular protein production. More preferably a cell line of the invention is provided wherein the cell line comprises a U-2 OS osteosarcoma, CHO, 293, HuNS-1 myeloma, WERI-Rb-1 retinoblastoma, BHK, Vero, non-secreting mouse myeloma Sp2/0-Ag 14, non-secreting mouse myeloma NSO, or NCI-H295R adrenal gland carcinoma (ATCC CRL-2128) cell line.
A cell line of the invention is particularly suitable for production of a proteinaceous molecule, because production can be enhanced by one or more STAR sequences (higher yield of a proteinaceous molecule, higher proportion of host cells with acceptable expression levels, and/or higher stability of a gene expression level). A cell line of the invention can comprise promiscuous, species-specific and/or cell type-specific STAR sequences. Furthermore, a cell line of the invention can be used to produce a heterologous proteinaceous molecule, and/or an affiliated proteinaceous molecule.
Hence, a use of a cell line of the invention for the production of a proteinaceous molecule is also herewith provided. Preferably, the proteinaceous molecule comprises an affiliated protein of the cell line.
Of course, a proteinaceous molecule obtainable by a method of the invention is also provided by the present invention.
In one aspect, the invention provides a method for selecting a cell suitable for producing a proteinaceous molecule comprising:
    • providing a nucleic acid encoding the proteinaceous molecule with a nucleic acid comprising a STAR sequence;
    • expressing the resulting nucleic acid in the cell; and
    • determining whether produced proteinaceous molecule has a desired property.
The desired property, for instance, comprises a pharmaceutical property. The property can be influenced by post translational modification(s), a configuration of a produced proteinaceous molecule, etc.
In yet another aspect, the invention provides a method for selecting a cell suitable for producing a proteinaceous molecule comprising:
    • providing a host cell with a nucleic acid comprising a STAR sequence;
    • selecting a cell with enhanced expression of a proteinaceous molecule; and
    • determining whether the proteinaceous molecule has a desired property.
As has been discussed above, the nucleic acid comprising a STAR sequence can be randomly introduced into the genome of the host cell. Preferably, however, the nucleic acid sequence is introduced into the genome of the host cell by homologous recombination.
The invention is further explained in the following examples. The examples do not limit the invention in any way. They merely serve to clarify the invention.
EXAMPLES Example 1 Method for Isolation of Star Elements from the Human Genome
STAR elements are identified and cloned from human genomic DNA based on their ability to block the spread of transcriptional repression from DNA binding sites for repressor proteins in a test vector, as described in this example. The method described in this example is applicable in principle to any mammalian cell line, for isolation of both promiscuous and cell line-specific STAR elements.
A Method to Isolate Human Star Elements Functional in U-2 Os Cell
Materials and Methods
Plasmids and strains. The selection vector for STAR elements, pSelect-SV40-zeo (“pSelect,” FIG. 1) was constructed as follows: the pREP4 vector (Invitrogen V004-50) was used as the plasmid backbone. It provides the Epstein Barr oriP origin of replication and EBNA-1 nuclear antigen for high-copy episomal replication in primate cell lines; the hygromycin resistance gene with the thymidine kinase promoter and polyadenylation site, for selection in mammalian cells; and the ampicillin resistance gene and colE1 origin of replication for maintenance in Escherichia coli. The vector contains four consecutive LexA operator sites between XbaI and NheI restriction sites (Bunker and Kingston, 1994). Embedded between the LexA operators and the NheI site is a polylinker consisting of the following restriction sites: HindIII-AscI-BamHI-AscI-HindIII. Between the NheI site and a SalI site is the zeocin resistance gene with the SV40 promoter and polyadenylation site, derived from pSV40/Zeo (Invitrogen V502-20); this is the selectable marker for the STAR screen.
Gene libraries were constructed by Sau3AI digestion of human genomic DNA, either purified from placenta (Clontech 6550-1) or carried in bacterial/P1 (BAC/PAC) artificial chromosomes. The BAC/PAC clones contain genomic DNA from the 1q12 cytogenetic region (clones RP1154H19 and RP3328E19), from the HOX cluster of homeotic genes (clones RP1167F23, RP1170019, and RP11387A1), or from human chromosome 22 (Research Genetics 96010-22). The DNAs were size-fractionated, and the 0.5-2 kb size fraction ligated into BamHI-digested pSelect vector, by standard techniques (Sambrook et al., 1989).
The construction of the host strains has been described (van der Vlag et al., 2000). Briefly, they are based on the U-2 OS human osteosarcoma cell line (American Type Culture Collection HTB-96). U-2 OS was stably transfected with the pTet-Off plasmid (Clontech K1620-A), encoding a protein chimera consisting of the Tet-repressor DNA binding domain and the VP16 transactivation domain. The cell line was subsequently stably transfected with fusion protein genes containing the LexA DNA binding domain, and the coding regions of HP1, MeCP2, or HPC2 (three Drosophila proteins that repress gene expression when tethered to DNA). The LexA-repressor genes are under control of the Tet-Off transcriptional regulatory system (Gossen and Bujard, 1992).
Library screening and STAR element characterization. The gene libraries in pSelect were transfected into U-2 OS/Tet-Off/LexA-repressor cell lines by calcium phosphate precipitation (Graham and van der Eb, 1973, Wigler et al., 1978) as recommended by the supplier of the transfection reagent (Life Technologies). Transfected cells were cultured under hygromycin selection (25 μg/ml) and tetracycline repression (doxycycline, 10 ng/ml) for one week (50% confluence). Then the doxycycline concentration was reduced to 0.1 ng/ml to induce the LexA-repressor genes, and after two days zeocin was added to 250 μg/ml. The cells are cultured for a further four to five weeks, until the control cultures (transfected with empty pSelect) were killed by the zeocin.
Zeocin-resistant colonies from the library transfection were propagated, and plasmid DNA isolated and rescued into E. coli by standard techniques (Sambrook et al., 1989). The candidate STAR elements in the rescued DNA were analyzed by restriction endonuclease mapping (Sambrook et al., 1989), and tested for STAR activity (zeocin resistance) after re-transfection to U-2 OS/Tet-Off/LexA-repressor cells and lowering the doxycycline concentration.
The human genomic DNA inserts in these plasmids were sequenced by the dideoxy method (Sanger et al., 1977) using a Beckman CEQ™2000 automated DNA sequencer, using the manufacturer's instructions. Briefly, DNA was purified from E. coli using QIAprep® Spin Miniprep and Plasmid Midi Kits (QIAGEN® 27106 and 12145, respectively). Cycle sequencing was carried out using custom oligonucleotides corresponding to the pSelect vector (primers D89 (SEQ ID NO:149) and D95 (SEQ ID NO:154); all oligonucleotides are described in Table 2), in the presence of dye terminators (CEQ™ Dye Terminator Cycle Sequencing Kit, Beckman 608000). Assembled STAR DNA sequences were located in the human genome using BLAST (Basic Local Alignment Search Tool (Altschul et al., 1990); worldwideweb.ncbi.nlm.nih.gov/BLAST/).
Results
The screens of human genomic DNA have yielded 66 STAR elements; the lengths and chromosomal locations of these elements are tabulated in SEQ ID NO:1-SEQ ID NO:66. They confer zeocin resistance on U-2 OS host cells when placed between LexA-repressor binding sites and the zeocin resistance gene. Their anti-repression activity was demonstrated both in the initial screen and upon re-transfection (demonstrating that the anti-repression activity is due to the STAR element and not to somatic acquisition of zeocin resistance). The STAR elements correspond to known and unique sequences in the human genome, as demonstrated by BLAST searches (Table 3). In some cases, the cloned element is a chimera of two unlinked genomic loci (e.g., STAR3 (SEQ ID NO:3), Table 3). They range in length from 500 to 2361 base pairs in length.
Example 2 Predictability and Yield is Improved by Promiscuous Star Elements in More than One Host Cell Line
STAR elements function to block the effect of transcriptional repression influences on transgene expression units. These repression influences can be due to heterochromatin (“position effects”) or to adjacent copies of the transgene (“repeat-induced gene silencing”). Two of the benefits of STAR elements for heterologous protein production are increased predictability of finding high-expressing primary recombinant host cells and increased yield during production cycles. These benefits are illustrated in this example.
Materials and Methods
Construction of the pSDH vectors and STAR-containing derivatives: The pSDH-Tet vector was constructed by polymerase chain reaction amplification (PCR) of the luciferase open reading frame from plasmid pREP4-HSF-Luc (van der Vlag et al., 2000) using primers C67 (SEQ ID NO:136) and C68 (SEQ ID NO:137), and insertion of the SacII/BamHI fragment into SacII/BamHI-digested pUHD10-3 (Gossen and Bujard, 1992). The luciferase expression unit was re-amplified with primers C65 (SEQ ID NO:134) and C66 (SEQ ID NO:135), and re-inserted into pUHD10-3 in order to flank it with multiple cloning sites (MCSI and MCSII). An AscI site was then introduced into MCSI by digestion with EcoRI and insertion of a linker (comprised of annealed oligonucleotides D93 (SEQ ID NO:152) and D94 (SEQ ID NO:153)). The CMV promoter was amplified from plasmid pCMV-Bsd (Invitrogen K510-01) with primers D90 (SEQ ID NO:150) and D91 (SEQ ID NO:151), and used to replace the Tet-Off promoter in pSDH-Tet by SalI/SacII digestion and ligation to create vector pSDH-CMV. The luciferase open reading frame in this vector was replaced by SEAP (Secreted Alkaline Phosphatase) as follows: vector pSDH-CMV was digested with SacII and BamHI and made blunt; the SEAP open reading frame was isolated from pSEAP-basic (Clontech 6037-1) by EcoRI/SalI digestion, made blunt and ligated into pSDH-CMV to create vector pSDH-CS. The puromycin resistance gene under control of the SV40 promoter was isolated from plasmid pBabe-Puro (Morgenstern and Land, 1990) by PCR, using primers C81 (SEQ ID NO:138) and C82 (SEQ ID NO:139). This was ligated into vector pGL3-control (BamHI site removed) (Promega E1741) digested with NcoI/XbaI to create pGL3-puro. pGL3-puro was digested with BglII/SalI to isolate the SV40-puro resistance unit, which was made blunt and ligated into NheI digested, blunt-ended pSDH-CS. The resulting vector, pSDH-CSP, is shown in FIG. 2. All cloning steps were carried out following the instructions provided by the manufacturers of the reagents used, according to methods known in the art (Sambrook et al., 1989).
STAR elements were inserted into MCSI and MCSII in two steps, by digestion of the STAR element and the pSDH-CSP vector with an appropriate restriction enzyme, followed by ligation. The orientation of STAR elements in recombinant pSDH vectors was determined by restriction mapping, and in all cases verified by DNA sequence analysis using primers C85 (SEQ ID NO:140), E42 (SEQ ID NO:168), and E25 (SEQ ID NO:167) (Table 2; see Example 1).
Transfection and culture of U-2 OS cells with pSDH-CMV plasmids: The human osteosarcoma U-2 OS cell line (ATCC #HTB-96) was cultured in Dulbecco's Modified Eagle Medium+10% Fetal Calf Serum containing glutamine, penicillin, and streptomycin (supra) at 37° C./5% CO2. Cells were co-transfected with the pSDH-CMV vector and its derivatives containing STAR6 (SEQ ID NO:6) or STAR8 (SEQ ID NO:8) in MCSI and MCSII (along with plasmid pBabe-Puro) using SuperFect® (supra). Puromycin selection was complete in two weeks, after which time individual puromycin resistant U-2 OS/pSDH-CMV clones were isolated at random and cultured further.
Luciferase assay: Luciferase activity (Himes and Shannon, 2000) was assayed in resuspended cells according to the instructions of the assay kit manufacturer (Roche 1669893), using a luminometer (Turner 20/20TD). Total cellular protein concentration was determined by the bicinchoninic acid method according to the manufacturer's instructions (Sigma B-9643), and used to normalize the luciferase data.
Transfection and culture of CHO cells with pSDH-CSP plasmids: The Chinese Hamster Ovary cell line CHO-K1 (ATCC CCL-61) was cultured in HAMS-F12 medium+10% Fetal Calf Serum containing 2 mM glutamine, 100 U/ml penicillin, and 100 micrograms/ml streptomycin at 37° C./5% CO2. Cells were transfected with recombinant pSDH-CSP vectors using SuperFect® (QIAGEN®) as described by the manufacturer. Briefly, cells were seeded to culture vessels and grown overnight to 70-90% confluence. SuperFect® reagent was combined with plasmid DNA (linearized in this example by digestion with PvuI) at a ratio of 6 microliters per microgram (e.g., for a 10 cm Petri dish, 20 micrograms DNA and 120 microliters SuperFect®) and added to the cells. After overnight incubation, the transfection mixture was replaced with fresh medium, and the transfected cells were incubated further. After overnight cultivation, 5 micrograms/ml puromycin was added. Puromycin selection was complete in two weeks, after which time individual puromycin resistant CHO/pSDH-CSP clones were isolated at random and cultured further.
Secreted Alkaline Phosphatase (SEAP) assay: SEAP activity (Berger et al., 1988, Henthorn et al., 1988, Kain, 1997, Yang et al., 1997) in the culture medium of CHO/pSDH-CSP clones was determined as described by the manufacturer (Clontech Great EscAPe kit #K2041). Briefly, an aliquot of medium was heat inactivated at 65° C., then combined with assay buffer and CSPD chemiluminescent substrate and incubated at room temperature for ten minutes. The rate of substrate conversion was then determined in a luminometer (Turner 20/20TD). Cell density was determined by counting trypsinized cells in a Coulter ACT10 cell counter. Luminescence units were converted into picograms SEAP based on a SEAP positive control calibration curve, and normalized to cell number.
Results
Recombinant U-2 OS cell clones containing the pSDH-CMV vector, or a pSDH-CMV plasmid containing STAR6 (SEQ ID NO:6) (Table 3), were cultured for three weeks. The luciferase activity in the host cells was then determined, and is expressed as relative luciferase units (FIG. 3), normalized to total cell protein. The recombinant U-2 OS clones with STAR6 (SEQ ID NO:6) flanking the expression units had higher yields than the STAR-less clones: the STAR6 clones had maximal luciferase expression levels five-fold higher than the STAR-less clones. The STAR6 (SEQ ID NO:6) element conferred greater predictability as well: 15-20% of the clones expressed luciferase at levels comparable to or greater than the STAR-less clone with the highest expression level.
Recombinant CHO cell clones containing the pSDH-CSP vector, or a pSDH-CSP plasmid containing STAR6 (SEQ ID NO:6), were cultured for three weeks. The SEAP activity in the culture supernatants was then determined, and is expressed on the basis of cell number (FIG. 4). As can be seen, clones with the STAR6 (SEQ ID NO:6) element in the expression units were isolated that express two- to three-fold higher SEAP activity than clones whose expression units do not include this STAR element. Furthermore, the number of STAR6-containing (SEQ ID NO:6-containing) clones that express SEAP activity at or above the maximal activity of the STAR-less clones is quite high: 40% of the STAR6 clone populations exceed the highest SEAP expression of the pSDH-CSP clones.
These results demonstrate that, when used with the strong CMV promoter, the STAR6 (SEQ ID NO:6) element increases the yield of this heterologous protein in both of the host cell lines tested. STAR6 (SEQ ID NO:6) also confers increased predictability, as manifested by the large proportion of the clones with yields equal to or greater than the highest yield displayed by the STAR-less clones. Thus, STAR6 (SEQ ID NO:6) is an example of a promiscuous STAR element, able to suppress transgene repression in more than one host cell line. The cell lines used are derived from different species (human and hamster) and different tissue types (bone and ovary), reflecting the broad range of host cells in which this STAR element can be utilized in improving heterologous protein expression.
Example 3 STAR8 (SEQ ID NO:8) is a Cell Line-Specific STAR Element
The patterns of gene expression and epigenetic gene regulation in a host cell line reflect the developmental state of the somatic cells from which they are derived. Furthermore, the biotechnology industry takes advantage of general purpose cell lines from different species according to specific requirements of a heterologous protein production process. Therefore, it is expected that some STAR elements will not function in cell lines other than those in which they are isolated. This expectation has been fulfilled by some of the STAR elements shown in Table 3. One example will be given here.
Materials and Methods
pSDH vector construction, transfection and cultivation of CHO and U-2 OS cell lines, and assay methods for the SEAP and luciferase reporter genes has been described in Example 2.
Results
Recombinant U-2 OS cell clones containing the pSDH-CMV vector, or a pSDH-CMV plasmid containing STAR8 (SEQ ID NO:8) (Table 3), were cultured for three weeks. The luciferase activity in the host cells was then determined, and is expressed as relative luciferase units (FIG. 3), normalized to total cell protein. The recombinant U-2 OS clones with the STAR8 (SEQ ID NO:8) element flanking the expression units had higher yields than the STAR-less clones: the highest expression observed from STAR8 clones was two- to three-fold higher than the expression from STAR-less clones. The STAR8 (SEQ ID NO:8) element conferred greater predictability as well: for this STAR element, ˜15% of the clones displayed luciferase expression at levels comparable to or greater than the STAR-less clone with the highest expression level.
Recombinant CHO cell clones transfected with the pSDH-CSP vector, or a pSDH-CSP plasmid containing STAR8 (SEQ ID NO:8), were cultured for three weeks. The SEAP activity in the culture supernatants was then determined, and is expressed on the basis of cell number (FIG. 4). As can be seen, one clone with the STAR8 (SEQ ID NO:8) element in the expression unit had a yield approximately two-fold higher than the highest-expressing STAR-less clone. However, the rest of the STAR8 clones expressed very poorly relative to the STAR-less clone population. Since only one individual in the STAR8 population had a good yield, it is probable that the expression unit in this clone was integrated in open, transcriptionally active chromatin, and the high yield does not reflect anti-repression activity of STAR8 (SEQ ID NO:8) in CHO cells. Certainly in the CHO clones transfected with STAR8-containing (SEQ ID NO:8-containing) expression units the predictability is quite poor; of the 17 puromycin-resistant clones, only one clone had a yield of SEAP activity above the background level of expression.
This example demonstrates that good performance of a STAR element in one cell line (in this case, the U-2 OS cell line in which STAR8 (SEQ ID NO:8) was originally isolated) is not an accurate predictor of its performance in other cell lines. STAR8 (SEQ ID NO:8) is thus an example of a cell line-specific STAR element.
Example 4 STAR Elements Functionality in Diverse Cell Line
Materials and Methods
Cell lines including the U-2 OS osteosarcoma and CHO (Chinese hamster ovary) cell lines (supra), the 293 cell line (ATCC CRL-1573) derived from human embryonal kidney (immortalized by adenovirus 5 transfection), the HuNS-1 myeloma (ATCC CRL-8644) and the WERI-Rb-1 retinoblastoma cell line (ATCC HTB-169), the NCI-H295R adrenal gland carcinoma (ATCC CRL-2128), and the non-secreting mouse myelomas Sp2/0-Ag 14 and NSO are examined according to the previous examples.
Example 5 STAR Elements Improve the Stability of Transgene Expression
During cultivation of recombinant host cells, it is common practice to maintain antibiotic selection. This is intended to prevent transcriptional silencing of the transgene, or loss of the transgene from the genome by processes such as recombination. However it is undesirable for production of heterologous proteins, for a number of reasons. First, the antibiotics that are used are quite expensive, and contribute significantly to the unit cost of the product. Second, for biopharmaceutical use, the protein must be demonstrably pure, with no traces of the antibiotic in the product. One advantage of STAR elements for heterologous protein production is that they confer stable expression on transgenes during prolonged cultivation, even in the absence of antibiotic selection; this property is demonstrated in this example.
Materials and Methods
The U-2 OS cell line was transfected with the plasmid pSDH-Tet-STAR6 and cultivated as described in Example 2. Individual puromycin-resistant clones were isolated and cultivated further in the absence of doxycycline. At weekly intervals the cells were transferred to fresh culture vessels at a dilution of 1:20. Luciferase activity was measured at periodic intervals as described in Example 2. After 15 weeks, the cultures were divided into two replicates; one replicate continued to receive puromycin, while the other replicate received no antibiotic for the remainder of the experiment (25 weeks total).
Results
Table 4 presents the data on luciferase expression by an expression unit flanked with STAR6 (SEQ ID NO:6) during prolonged growth with or without antibiotic. As can be seen, the expression of the reporter transgene, luciferase, remains stable in the U-2 OS host cells for the duration of the experiment. After the cultures were divided into two treatments (plus antibiotic and without antibiotic) the expression of luciferase was essentially stable in the absence of antibiotic selection. This demonstrates the ability of STAR elements to protect transgenes from silencing or loss during prolonged cultivation. It also demonstrates that this property is independent of antibiotic selection. Therefore, production of heterologous proteins is possible without incurring the costs of the antibiotic or of difficult downstream processing.
Example 6 Minimal Essential Sequences of STAR Elements
STAR elements are isolated from the genetic screen described in Example 1. The screen uses libraries constructed with human genomic DNA that was size-fractionated to approximately 0.5-2 kilobases (supra). The STAR elements range from 500 to 2361 base pairs (Table 3). It is likely that, for many of the STAR elements that have been isolated, STAR activity is conferred by a smaller DNA fragment than the initially isolated clone. It is useful to determine these minimum fragment sizes that are essential for STAR activity, for two reasons. First, smaller functional STAR elements would be advantageous in the design of compact expression vectors, since smaller vectors transfect host cells with higher efficiency. Second, determining minimum essential STAR sequences permits the modification of those sequences for enhanced functionality. Two STAR elements have been fine-mapped to determine their minimal essential sequences.
Materials and Methods
STAR10 (SEQ ID NO:10) (1167 base pairs) and STAR27 (SEQ ID NO:27) (1520 base pairs) have been fine-mapped. They have been amplified by PCR to yield sub-fragments of approximately equal length (FIG. 6 legend). For initial testing, these have been cloned into the pSelect vector at the BamHI site, and transfected into U-2 OS/Tet-Off/LexA-HP1 cells as described in Example 1. After selection for hygromycin resistance, LexA-HP1 was induced by lowering the doxycycline concentration. Transfected cells were then incubated with zeocin to test the ability of the STAR fragments to protect the SV40-Zeo expression unit from repression due to LexA-HP1 binding.
Results
In this experiment STAR10 (SEQ ID NO:10) and STAR 27 (SEQ ID NO:27) confer good protection against gene silencing, as expected (FIG. 6). This is manifested by robust growth in the presence of zeocin.
Of the three STAR10 (SEQ ID NO:10) sub-fragments, 10A (˜400 base pairs, corresponding to approximately the first 400 nucleotides of SEQ ID NO:10) confers on transfected cells vigorous growth in the presence of zeocin, exceeding that of the full-length STAR element. Cells transfected with pSelect constructs containing the other two sub-fragments do not grow in the presence of zeocin. These results identify the ˜400 base pair 10A fragment as encompassing the DNA sequence responsible for the anti-repression activity of STAR10 (SEQ ID NO:10).
STAR27 (SEQ ID NO:27) confers moderate growth in zeocin to transfected cells in this experiment (FIG. 6). One of the sub-fragments of this STAR, 27B (˜500 base pairs, corresponding to approximately the second 500 nucleotides of SEQ ID NO:27), permits weak growth of the host cells in zeocin-containing medium. This suggests that the anti-repression activity of this STAR is partially localized on sub-fragment 27B, but full activity requires sequences from 27A (corresponding to approximately the first 500 nucleotides of SEQ ID NO:27) and/or 27C (corresponding to approximately the third 500 nucleotides of SEQ ID NO:27) (each ˜500 base pairs) as well.
Example 7 STAR Elements Function in the Context of Various Transcriptional Promoters
Transgene transcription is achieved by placing the transgene open reading frame under control of an exogenous promoter. The choice of promoter is influenced by the nature of the heterologous protein and the production system. In most cases, strong constitutive promoters are preferred because of the high yields they can provide. Some viral promoters have these properties; the promoter/enhancer of the cytomegalovirus immediate early gene (“CMV promoter”) is generally regarded as the strongest promoter in common biotechnological use (Boshart et al., 1985, Doll et al., 1996, Foecking and Hofstetter, 1986). The simian virus SV40 promoter is also moderately strong (Boshart et al., 1985, Foecking and Hofstetter, 1986) and is frequently used for ectopic expression in mammalian cell vectors. The Tet-Off promoter is inducible: the promoter is repressed in the presence of tetracycline or related antibiotics (doxycycline is commonly used) in cell-lines which express the tTA plasmid (Clontech K1620-A), and removal of the antibiotic results in transcriptional induction (Deuschle et al., 1995, Gossen and Bujard, 1992, Izumi and Gilbert, 1999, Umana et al., 1999).
Materials and Methods
The construction of the pSDH-Tet and pSDH-CMV vectors is described in Example 2. pSDH-SV40 was constructed by PCR amplification of the SV40 promoter (primers D41 (SEQ ID NO:142) and D42 (SEQ ID NO:143)) from plasmid pSelect-SV40-Zeo (Example 1), followed by digestion of the PCR product with SacII and SalI. The pSDH-CMV vector was digested with SacII and SalI to remove the CMV promoter, and the vector and SV40 fragment were ligated together to create pSDH-SV40. STAR6 (SEQ ID NO:6) was cloned into MCSI and MCSII as described in Example 2. The plasmids pSDH-Tet, pSDH-Tet-STAR6, pSDH-Tet-STAR7, pSDH-SV40 and pSDH-SV40-STAR6 were co-transfected with pBabe-Puro into U-2 OS using SuperFect® as described by the manufacturer. Cell cultivation, puromycin selection, and luciferase assays were carried out as described in Example 2.
Results
FIGS. 3, 7, and 8 compare the expression of the luciferase reporter gene from three different promoters: two strong and constitutive viral promoters (CMV and SV40), and the inducible Tet-Off promoter. All three promoters were tested in the context of the STAR6 (SEQ ID NO:6) element in U-2 OS cells. The results demonstrate that the yield and predictability from all three promoters are increased by STAR6 (SEQ ID NO:6). As described in Examples 2 and 5, STAR6 (SEQ ID NO:6) is beneficial in the context of the CMV promoter (FIG. 3). Similar improvements are seen in the context of the SV40 promoter (FIG. 7): the yield from the highest-expressing STAR6 clone is two- to three-fold greater than the best pSDH-SV40 clones, and six STAR clones (20% of the population) have yields higher than the best STAR-less clones. In the context of the Tet-Off promoter under inducing (low doxycycline) concentrations, STAR6 (SEQ ID NO:6) also improves the yield and predictability of transgene expression (FIG. 8): the highest-expressing STAR6 clone has a 20-fold higher yield than the best pSDH-Tet clone, and nine STAR6 clones (35% of the population) have yields higher than the best STAR-less clone. It is concluded that this STAR element is versatile in its transgene-protecting properties, since it functions in the context of various biotechnologically useful promoters of transcription.
Example 8 STAR Element Function can be Directional
While short nucleic acid sequences can be symmetrical (e.g., palindromic), longer, naturally-occurring sequences are typically asymmetrical. As a result, the information content of nucleic acid sequences is directional and the sequences themselves can be described with respect to their 5′ and 3′ ends. The directionality of nucleic acid sequence information affects the arrangement in which recombinant DNA molecules are assembled using standard cloning techniques known in the art (Sambrook et al., 1989). STAR elements are long, asymmetrical DNA sequences, and have a directionality based on the orientation in which they were originally cloned in the pSelect vector. In the examples given above, using two STAR elements in pSDH vectors, this directionality was preserved. This orientation is described as the native or 5′-3′ orientation, relative to the zeocin resistance gene (see FIG. 9). In this example the importance of directionality for STAR function is tested in the pSDH-Tet vector. Since the reporter genes in the pSDH vectors are flanked on both sides by copies of the STAR element of interest, the orientation of each STAR copy must be considered. This example compares the native orientation with the opposite orientation (FIG. 9).
Materials and Methods
The STAR66 (SEQ ID NO:66) element was cloned into pSDH-Tet as described in Example 2. U-2 OS cells were co-transfected with plasmids pSDH-Tet-STAR66-native and pSDH-Tet-STAR66-opposite, and cultivated as described in Example 2. Individual clones were isolated and cultivated; the level of luciferase expression was determined as described (supra).
Results
The results of the comparison of STAR66 (SEQ ID NO:66) activity in the native orientation and the opposite orientation are shown in FIG. 10. When STAR66 (SEQ ID NO:66) is in the opposite orientation, the yield of only one clone is reasonably high (60 luciferase units). In contrast, the yield of the highest-expressing clone when STAR66 (SEQ ID NO:66) is in the native orientation is considerably higher (100 luciferase units) and the predictability is much higher, as well: seven clones of the native-orientation population (30%) express luciferase above the level of the highest-expressing clone from the opposite-orientation population, and 15 of the clones in the native-orientation population (60%) express luciferase above ten relative luciferase units. Therefore, it is demonstrated that STAR66 (SEQ ID NO:66) function is directional.
Example 9 Transgene Expression in the Context of STAR Elements is Copy Number-Dependent
Transgene expression units for heterologous protein expression are generally integrated into the genome of the host cell to ensure stable retention during cell division. Integration can result in one or multiple copies of the expression unit being inserted into the genome; multiple copies may or may not be present as tandem arrays. The increased yield demonstrated for transgenes protected by STAR elements (supra) suggests that STAR elements are able to permit the transgene expression units to function independently of influences on transcription associated with the site of integration in the genome (independence from position effects (Boivin and Dura, 1998)). It suggests further that the STAR elements permit each expression unit to function independently of neighboring copies of the expression unit when they are integrated as a tandem array (independence from repeat-induced gene silencing (Garrick et al., 1998)). Copy number-dependence is determined from the relationship between transgene expression levels and copy number, as described in the example below.
Materials and Methods
U-2 OS cells were co-transfected with pSDH-Tet-STAR10 and cultivated under puromycin selection as described (supra). Eight individual clones were isolated and cultivated further. Then cells were harvested, and one portion was assayed for luciferase activity as described (supra). The remaining cells were lysed and the genomic DNA purified using the DNeasy® Tissue Kit (QIAGEN® 69504) as described by the manufacturer. DNA samples were quantitated by UV spectrophotometry. Three micrograms of each genomic DNA sample were digested with PvuII and XhoI overnight as described by the manufacturer (New England Biolabs), and resolved by agarose gel electrophoresis. DNA fragments were transferred to a nylon membrane as described (Sambrook et al., 1989), and hybridized with a radioactively labeled probe to the luciferase gene (isolated from BamHI/SacII-digested pSDH-Tet). The blot was washed as described (Sambrook et al., 1989) and exposed to a phosphorimager screen (Personal F/X, BioRad). The resulting autoradiogram (FIG. 11) was analyzed by densitometry to determine the relative strength of the luciferase DNA bands, which represents the transgene copy number.
Results
The enzyme activities and copy numbers (DNA band intensities) of luciferase in the clones from the pSDH-Tet-STAR10 clone population is shown in FIG. 12. The transgene copy number is highly correlated with the level of luciferase expression in these pSDH-Tet-STAR10 clones (r=0.86). This suggests that STAR10 (SEQ ID NO:10) confers copy number-dependence on the transgene expression units, making transgene expression independent of other transgene copies in tandem arrays and independent of gene-silencing influences at the site of integration.
Example 10 STAR Elements Function as Enhancer Blockers but not Enhancers
Gene promoters are subject to both positive and negative influences on their ability to initiate transcription. An important class of elements that exert positive influences are enhancers. Enhancers are characteristically able to affect promoters even when they are located far away (many kilobase pairs) from the promoter. Negative influences that act by heterochromatin formation (e.g., Polycomb group proteins) have been described above, and these are the target of STAR activity. The biochemical basis for enhancer function and for heterochromatin formation is fundamentally similar, since they both involve binding of proteins to DNA. Therefore, it is important to determine whether STAR elements are able to block positive influences as well as negative influences, in other words, to shield transgenes from genomic enhancers in the vicinity of the site of integration. The ability to shield transgenes from enhancer activity ensures stable and predictable performance of transgenes in biotechnological applications. This example examines the performance of STAR elements in an enhancer-blocking assay.
Another feature of STAR activity that is important to their function is the increased yield they confer on transgenes (Example 2). STARs are isolated on the basis of their ability to maintain high levels of zeocin expression when heterochromatin-forming proteins are bound adjacent to the candidate STAR elements. High expression is predicted to occur because STARs are anticipated to block the spread of heterochromatin into the zeocin expression unit. However, a second scenario is that the DNA fragments in zeocin-resistant clones contain enhancers. Enhancers have been demonstrated to have the ability to overcome the repressive effects of Polycomb-group proteins such as those used in the method of the STAR screen (Zink and Paro, 1995). Enhancers isolated by this phenomenon would be considered false positives, since enhancers do not have the properties claimed here for STARs. In order to demonstrate that STAR elements are not enhancers, they have been tested in an enhancer assay.
The enhancer-blocking assay and the enhancer assay are methodologically and conceptually similar. The assays are shown schematically in FIG. 13. The ability of STAR elements to block enhancers is performed using the E47/E-box enhancer system. The E47 protein is able to activate transcription by promoters when it is bound to an E-box DNA sequence located in the vicinity of those promoters (Quong et al., 2002). E47 is normally involved in regulation of B and T lymphocyte differentiation (Quong et al., 2002), but it is able to function in diverse cell types when expressed ectopically (Petersson et al., 2002). The E-box is a palindromic DNA sequence, CANNTG (Knofler et al., 2002). In the enhancer-blocking assay, an E-box is placed upstream of a luciferase reporter gene (including a minimal promoter) in an expression vector. A cloning site for STAR elements is placed between the E-box and the promoter. The E47 protein is encoded on a second plasmid. The assay is performed by transfecting both the E47 plasmid and the luciferase expression vector into cells; the E47 protein is expressed and binds to the E-box, and the E47/E-box complex is able to act as an enhancer. When the luciferase expression vector does not contain a STAR element, the E47/E-box complex enhances luciferase expression (FIG. 13A, situation 1). When STAR elements are inserted between the E-box and the promoter, their ability to block the enhancer is demonstrated by reduced expression of luciferase activity (FIG. 13A, situation 2); if STARs cannot block enhancers, luciferase expression is activated (FIG. 13A, situation 3).
The ability of STAR elements to act as enhancers utilizes the same luciferase expression vector. In the absence of E47, the E-box itself does not affect transcription. Instead, enhancer behavior by STAR elements will result in activation of luciferase transcription. The assay is performed by transfecting the luciferase expression vector without the E47 plasmid. When the expression vector does not contain STAR elements, luciferase expression is low (FIG. 13B, situation 1). If STAR elements do not have enhancer properties, luciferase expression is low when a STAR element is present in the vector (FIG. 13B, situation 2). If STAR elements do have enhancer properties, luciferase expression will be activated in the STAR-containing vectors (FIG. 13B, situation 3).
Materials and Methods
The luciferase expression vector was constructed by inserting the E-box and a human alkaline phosphatase minimal promoter from plasmid mu-E5+E2×6-cat(x) (Ruezinsky et al., 1991) upstream of the luciferase gene in plasmid pGL3-basic (Promega E1751), to create pGL3-E-box-luciferase (gift of W. Romanow). The E47 expression plasmid contains the E47 open reading frame under control of a beta-actin promoter in the pHBAPr-1-neo plasmid; E47 in constitutively expressed from this plasmid (gift of W. Romanow).
STAR elements 1, 2, 3, 6, 10, 11, 18, and 27 (SEQ ID NOS:1, 2, 3, 6, 10, 11, 18, and 27, respectively) have been cloned into the luciferase expression vector. Clones containing the Drosophila scs element and the chicken beta-globin HS4-6× core (“HS4”) element have been included as positive controls (they are known to block enhancers, and to have no intrinsic enhancer properties (Chung et al., 1993, Kellum and Schedl, 1992)), and the empty luciferase expression vector has been included as a negative control. All assays were performed using the U-2 OS cell line. In the enhancer-blocking assay, the E47 plasmid was co-transfected with the luciferase expression vectors (empty vector, or containing STAR or positive-control elements). In the enhancer assay, the E47 plasmid was co-transfected with STARless luciferase expression vector as a positive control for enhancer activity; all other samples received a mock plasmid during co-transfection. The transiently transfected cells were assayed for luciferase activity 48 hours after plasmid transfection (supra). The luciferase activity expressed from a plasmid containing no E-box or STAR/control elements was subtracted, and the luciferase activities were normalized to protein content as described (supra).
Results
FIG. 14 shows the results of the enhancer-blocking assay. In the absence of STAR elements (or the known enhancer-blocking elements scs and HS4), the E47/E-box enhancer complex activates expression of luciferase (“vector”); this enhanced level of expression has been normalized to 100. Enhancer activity is blocked by all STAR elements tested. Enhancer activity is also blocked by the HS4 and scs elements, as expected (Bell et al., 2001, Gerasimova and Corces, 2001). These results demonstrate that in addition to their ability to block the spreading of transcriptional silencing (negative influences), STAR elements are able to block the action of enhancers (positive influences).
FIG. 15 shows the results of the enhancer assay. The level of luciferase expression due to enhancement by the E47/E-box complex is set at 100 (“E47”). By comparison, none of the STAR elements bring about significant activation of luciferase expression. As expected, the scs and HS4 elements also do not bring about activation of the reporter gene. Therefore, it is concluded that at least the tested STAR elements do not possess enhancer properties.
Example 11 Star Elements are Conserved Between Mouse and Human
BLAT analysis of the STAR DNA sequence against the human genome database (http://genome.ucsc.edu/cgi-bin/hgGateway) reveals that some of these sequences have high sequence conservation with other regions of the human genome. These duplicated regions are candidate STAR elements; if they do show STAR activity, they would be considered paralogs of the cloned STARs (two genes or genetic elements are said to be paralogous if they are derived from a duplication event (Li, 1997)).
BLAST analysis of the human STARs against the mouse genome (http://www.ensembl.org/Mus_musculus/blastview) also reveals regions of high sequence conservation between mouse and human. This sequence conservation has been shown for fragments of 15 out of the 65 human STAR elements. The conservation ranges from 64% to 89%, over lengths of 141 base pairs to 909 base pairs (Table 5). These degrees of sequence conservation are remarkable and suggest that these DNA sequences may confer STAR activity within the mouse genome as well. Some of the sequences from the mouse and human genomes in Table 5 could be strictly defined as orthologs (two genes or genetic elements are said to be orthologous if they are derived from a speciation event (Li, 1997)). For example, STAR6 (SEQ ID NO:6) is between the SLC8A1 and HAAO genes in both the human and mouse genomes. In other cases, a cloned human STAR has a paralog within the human genome, and its ortholog has been identified in the mouse genome. For example, STAR3a is a fragment of the 15q11.2 region of human chromosome 15. This region is 96.9% identical (paralogous) with a DNA fragment at 5q33.3 on human chromosome 5, which is near the IL12B interleukin gene. These human DNAs share approximately 80% identity with a fragment of the 11B2 region on mouse chromosome 11. The 11B2 fragment is also near the (mouse) IL12B interleukin gene. Therefore, STAR3a and the mouse 11B2 fragment can be strictly defined as paralogs.
In order to test the hypothesis that STAR activity is shared between regions of high sequence conservation in the mouse and human genome, one of the human STARs with a conserved sequence in mouse, STAR18 (SEQ ID NO:18), has been analyzed in greater detail. The sequence conservation in the mouse genome detected with the original STAR18 clone extends leftward on human chromosome 2 for about 500 base pairs (FIG. 16; left and right relate to the standard description of the arms of chromosome 2). In this example, we examine whether the region of sequence conservation defines a “naturally occurring” STAR element in human that is more extensive in length than the original clone. We also examine whether the STAR function of this STAR element is conserved between mouse and human.
Materials and Methods
The region of mouse/human sequence conservation around STAR18 (SEQ ID NO:18) was recovered from human BAC clone RP11-387A1 by PCR amplification, in three fragments: the entire region (primers E93 (SEQ ID NO:171) and E94 (SEQ ID NO:172)), the leftward half (primers E93 (SEQ ID NO:171) and E92 (SEQ ID NO:170)), and the rightward half (primers E57 (SEQ ID NO:169) and E94 (SEQ ID NO:172)). The corresponding fragments from the homologous mouse region were recovered from BAC clone RP23-400H17 in the same fashion (primers E95 (SEQ ID NO:173) and E98 (SEQ ID NO:176), E95 (SEQ ID NO:173) and E96 (SEQ ID NO:174), and E97 (SEQ ID NO:175) and E98 (SEQ ID NO:176), respectively). All fragments were cloned into the pSelect vector and transfected into a U-2 OS/Tet-Off/LexA-HP1 cell line (supra). Following transfection, hygromycin selection was carried out to select for transfected cells. The LexA-HP1 protein was induced by lowering the doxycycline concentration, and the ability of the transfected cells to withstand the antibiotic zeocin (a measure of STAR activity) was assessed by monitoring cell growth.
Results
The original STAR18 clone was isolated from Sau3AI digested human DNA ligated into the pSelect vector on the basis of its ability to prevent silencing of a zeocin resistance gene. Alignment of the human STAR18 clone (497 base pairs) with the mouse genome revealed high sequence similarity (72%) between the orthologous human and mouse STAR18 (SEQ ID NO:18) regions. It also uncovered high similarity (73%) in the region extending for 488 base pairs immediately to the left of the Sau3AI site that defines the left end of the cloned region (FIG. 16). Outside these regions the sequence similarity between human and mouse DNA drops below 60%.
As indicated in FIG. 16, both the human and the mouse STAR18 (SEQ ID NO:18) elements confer survival on zeocin to host cells expressing the lexA-HP1 repressor protein. The original 497 base pair STAR18 clone and its mouse ortholog both confer the ability to grow (FIG. 16, a and d). The adjacent 488 base pair regions of high similarity from both genomes also confer the ability to grow, and in fact their growth phenotype is more vigorous than that of the original STAR18 clone (FIG. 16, b and e). When the entire region of sequence similarity was tested, these DNAs from both mouse and human confer growth, and the growth phenotype is more vigorous than the two sub-fragments (FIG. 16, c and f). These results demonstrate that the STAR activity of human STAR18 (SEQ ID NO:18) is conserved in its ortholog from mouse. The high sequence conservation between these orthologous regions is particularly noteworthy because they are not protein-coding sequences, leading to the conclusion that they have some regulatory function that has prevented their evolutionary divergence through mutation.
This analysis demonstrates that cloned STAR elements identified by the original screening program may in some cases represent partial STAR elements, and that analysis of the genomic DNA in which they are embedded can identify sequences with stronger STAR activity.
Example 12 STAR Elements Contain Characteristic DNA Sequence Motifs
STAR elements are isolated on the basis of their anti-repression phenotype with respect to transgene expression. This anti-repression phenotype reflects underlying biochemical processes that regulate chromatin formation which are associated with the STAR elements. These processes are typically sequence-specific and result from protein binding or DNA structure. This suggests that STAR elements will share DNA sequence similarity. Identification of sequence similarity among STAR elements will provide sequence motifs that are characteristic of the elements that have already been identified by functional screens and tests. The sequence motifs will also be useful to recognize and claim new STAR elements whose functions conform to the claims of this patent. The functions include improved yield and stability of transgenes expressed in eukaryotic host cells.
Other benefits of identifying sequence motifs that characterize STAR elements include: (1) provision of search motifs for prediction and identification of new STAR elements in genome databases, (2) provision of a rationale for modification of the elements, and (3) provision of information for functional analysis of STAR activity. Using bio-informatics, sequence similarities among STAR elements have been identified; the results are presented in this example.
Bio-Informatic and Statistical Background
Regulatory DNA elements typically function via interaction with sequence-specific DNA-binding proteins. Bio-informatic analysis of DNA elements, such as STAR elements whose regulatory properties have been identified, but whose interacting proteins are unknown, requires a statistical approach for identification of sequence motifs. This can be achieved by a method that detects short DNA sequence patterns that are over-represented in a set of regulatory DNA elements (e.g., the STAR elements) compared to a reference sequence (e.g., the complete human genome). The method determines the number of observed and expected occurrences of the patterns in each regulatory element. The number of expected occurrences is calculated from the number of observed occurrences of each pattern in the reference sequence.
The DNA sequence patterns can be oligonucleotides of a given length, e.g., six base pairs. In the simplest analysis, for a six-base-pair oligonucleotide (hexamer) composed of the four nucleotides (A, C, G, and T) there are 46=4096 distinct oligonucleotides (all combinations from AAAAAA (SEQ ID NO:121) to TTTTTT (SEQ ID NO:122)). If the regulatory and reference sequences were completely random and had equal proportions of the A, C, G, and T nucleotides, then the expected frequency of each hexamer would be 1/4096 (˜0.00024). However, the actual frequency of each hexamer in the reference sequence is typically different than this due to biases in the content of G:C base pairs, etc. Therefore, the frequency of each oligonucleotide in the reference sequence is determined empirically by counting, to create a “frequency table” for the patterns.
The pattern frequency table of the reference sequence is then used to calculate the expected frequency of occurrence of each pattern in the regulatory element set. The expected frequencies are compared with the observed frequencies of occurrence of the patterns. Patterns that are “over-represented” in the set are identified; for example, if the hexamer ACGTGA (SEQ ID NO:123) is expected to occur five times in 20 kilobase pairs of sequence, but is observed to occur 15 times, then it is three-fold over-represented. Ten of the 15 occurrences of that hexameric sequence pattern would not be expected in the regulatory elements if the elements had the same hexamer composition as the entire genome. Once the over-represented patterns are identified, a statistical test is applied to determine whether their over-representation is significant, or may be due to chance. For this test, a significance index, “sig,” is calculated for each pattern. The significance index is derived from the probability of occurrence of each pattern, which is estimated by a binomial distribution. The probability takes into account the number of possible patterns (4096 for hexamers). The highest sig values correspond to the most overrepresented oligonucleotides (van Helden et al., 1998). In practical terms, oligonucleotides with sig ≧0 are considered as over-represented. A pattern with sig ≧0 is likely to be over-represented due to chance once (=100) in the set of regulatory element sequences. However, at sig ≧1 a pattern is expected to be over-represented once in ten (=101) sequence sets, sig ≧2 once in 100 (=102) sequence sets, etc.
The patterns that are significantly over-represented in the regulatory element set are used to develop a model for classification and prediction of regulatory element sequences. This employs Discriminant Analysis, a so-called “supervised” method of statistical classification known to one of ordinary skill in the art (Huberty, 1994). In Discriminant Analysis, sets of known or classified items (e.g., STAR elements) are used to “train” a model to recognize those items on the basis of specific variables (e.g., sequence patterns such as hexamers). The trained model is then used to predict whether other items should be classified as belonging to the set of known items (e.g., is a DNA sequence a STAR element). In this example, the known items in the training set are STAR elements (positive training set). They are contrasted with sequences that are randomly selected from the genome (negative training set) which have the same length as the STAR elements. Discriminant Analysis establishes criteria for discriminating positives from negatives based on a set of variables that distinguish the positives; in this example, the variables are the significantly over-represented patterns (e.g., hexamers).
When the number of over-represented patterns is high compared to the size of the training set, the model could become biased due to over-training. Over-training is circumvented by applying a forward stepwise selection of variables (Huberty, 1994). The goal of Stepwise Discriminant Analysis is to select the minimum number of variables that provides maximum discrimination between the positives and negatives. The model is trained by evaluating variables one-by-one for their ability to properly classify the items in the positive and negative training sets. This is done until addition of new variables to the model does not significantly increase the model's predictive power (i.e., until the classification error rate is minimized). This optimized model is then used for testing, in order to predict whether “new” items are positives or negatives (Huberty, 1994).
It is inherent in classification statistics that for complex items such as DNA sequences, some elements of the positive training set will be classified as negatives (false negatives), and some members of the negative training set will be classified as positives (false positives). When a trained model is applied to testing new items, the same types of misclassifications are expected to occur.
In the bio-informatic method described here, the first step, Pattern Frequency Analysis, reduces a large set of sequence patterns (e.g., all 4096 hexamers) to a smaller set of significantly over-represented patterns (e.g., 100 hexamers); in the second step, Stepwise Discriminant Analysis reduces the set of over-represented patterns to the subset of those patterns that have maximal discriminative power (e.g., five to ten hexamers). Therefore, this approach provides simple and robust criteria for identifying regulatory DNA elements such as STAR elements.
DNA-binding proteins can be distinguished on the basis of the type of binding site they occupy. Some recognize contiguous sequences; for this type of protein, patterns that are oligonucleotides of length six base pairs (hexamers) are fruitful for bio-informatic analysis (van Helden et al., 1998). Other proteins bind to sequence dyads: contact is made between pairs of highly conserved trinucleotides separated by a non-conserved region of fixed width (van Helden et al., 2000). In order to identify sequences in STAR elements that may be bound by dyad-binding proteins, frequency analysis was also conducted for this type of pattern, where the spacing between the two trinucleotides was varied from 0 to 20 (i.e., XXXN{0-20}XXX where X's are specific nucleotides composing the trinucleotides, and N's are random nucleotides from 0 to 20 base pairs in length). The results of dyad frequency analysis are also used for Linear Discriminant Analysis as described above.
Materials and Methods
Using the genetic screen described in the original patent application, sixty-six (66) STAR elements were initially isolated from human genomic DNA and characterized in detail (Table 3). The screen was performed on gene libraries constructed by Sau3AI digestion of human genomic DNA, either purified from placenta (Clontech 6550-1) or carried in bacterial/P1 (BAC/PAC) artificial chromosomes. The BAC/PAC clones contain genomic DNA from regions of chromosome 1 (clones RP1154H19 and RP3328E19), from the HOX cluster of homeotic genes (clones RP1167F23, RP1170019, and RP11387A1), or from human chromosome 22 (Research Genetics 96010-22). The DNAs were size-fractionated, and the 0.5-2 kb size fraction was ligated into BamHI-digested pSelect vector, by standard techniques (Sambrook et al., 1989). pSelect plasmids containing human genomic DNA that conferred resistance to zeocin at low doxycycline concentrations were isolated and propagated in Escherichia coli. The screens that yielded the STAR elements of Table 3 have assayed approximately 1-2% of the human genome.
The human genomic DNA inserts in these 66 plasmids were sequenced by the dideoxy method (Sanger et al., 1977) using a Beckman CEQ™2000 automated DNA sequencer, using the manufacturer's instructions. Briefly, DNA was purified from E. coli using QIAprep® Spin Miniprep and Plasmid Midi Kits (QIAGEN® 27106 and 12145, respectively). Cycle sequencing was carried out using custom oligonucleotides corresponding to the pSelect vector (primers D89 (SEQ ID NO:149) and D95 (SEQ ID NO:154), Table 2), in the presence of dye terminators (CEQ™ Dye Terminator Cycle Sequencing Kit, Beckman 608000). Assembled STAR DNA sequences were located in the human genome (database builds August and December 2001) using BLAT (Basic Local Alignment Tool (Kent, 2002); http://genome.ucsc.edu/cgi-bin/hgGateway; Table 3). In aggregate, the combined STAR sequences comprise 85.6 kilobase pairs, with an average length of 1.3 kilobase pairs.
Sequence motifs that distinguish STAR elements within human genomic DNA were identified by bio-informatic analysis using a two-step procedure, as follows (see FIG. 17 for a schematic diagram). The analysis has two input datasets: (1) the DNA sequences of the STAR elements (STAR1-STAR65 (SEQ ID NOS:1-65) were used; Table 3); and (2) the DNA sequence of the human genome (except for chromosome 1, which was not feasible to include due to its large size; for dyad analysis a random subset of human genomic DNA sequence (˜27 Mb) was used).
Pattern Frequency Analysis
The first step in the analysis uses RSA-Tools software (Regulatory Sequence Analysis Tools; http://www.ucmb.ulb.ac.be/bioinformatics/rsa-tools/; references (van Helden et al., 1998, van Helden et al., 2000, van Helden et al., 2000)) to determine the following information: (1) the frequencies of all dyads and hexameric oligonucleotides in the human genome; (2) the frequencies of the oligonucleotides and dyads in the 65 STAR elements; and (3) the significance indices of those oligonucleotides and dyads that are over-represented in the STAR elements compared to the genome. A control analysis was done with 65 sequences that were selected at random from the human genome (i.e., from 2689×103 kilobase pairs) that match the length of the STAR elements of Table 3.
Discriminant Analysis
The over-represented oligonucleotides and dyads were used to train models for prediction of STAR elements by Linear Discriminant Analysis (Huberty, 1994). A pre-selection of variables was performed by selecting the 50 patterns with the highest individual discriminatory power from the over-represented oligos or dyads of the frequency analyses. These pre-selected variables were then used for model training in a Stepwise Linear Discriminant Analysis to select the most discriminant combination of variables (Huberty, 1994). Variable selection was based on minimizing the classification error rate (percentage of false negative classifications). In addition, the expected error rate was estimated by applying the same discriminant approach to the control set of random sequences (minimizing the percentage of false positive classifications).
The predictive models from the training phase of Discriminant Analysis were tested in two ways. First, the STAR elements and random sequences that were used to generate the model (the training sets) were classified. Second, sequences in a collection of 19 candidate STAR elements (recently cloned by zeocin selection as described above) were classified. These candidate STAR elements are listed in Table 8 (SEQ ID NOS:66-84).
Results
Pattern frequency analysis was performed with RSA-Tools on 65 STAR elements, using the human genome as the reference sequence. One hundred sixty-six (166) hexameric oligonucleotides were found to be over-represented in the set of STAR elements (sig ≧0) compared to the entire genome (Table 6). The most significantly over-represented oligonucleotide, CCCCAC (SEQ ID NO:177), occurs 107 times among the 65 STAR elements, but is expected to occur only 49 times. It has a significance coefficient of 8.76; in other words, the probability that its over-representation is due to random chance is 1/108.76, i.e., less than one in 500 million.
Ninety-five of the oligonucleotides have a significance coefficient greater than one, and are, therefore, highly over-represented in the STAR elements. Among the over-represented oligonucleotides, their observed and expected occurrences, respectively, range from 6 and 1 (for oligo 163, CGCGAA (SEQ ID NO:339), sig=0.02) to 133 and 95 (for oligo 120, CCCAGG (SEQ ID NO:296), sig=0.49). The differences in expected occurrences reflect factors such as the G:C content of the human genome. Therefore, the differences among the oligonucleotides in their number of occurrences is less important than their over-representation; for example, oligo 2 (CAGCGG (SEQ ID NO:178)) is 36/9=four-fold over-represented, which has a probability of being due to random chance of one in fifty million (sig=7.75).
Table 6 also presents the number of STAR elements in which each over-represented oligonucleotide is found. For example, the most significant oligonucleotide, oligo 1 (CCCCAC (SEQ ID NO:177)), occurs 107 times, but is found in only 51 STARs, i.e., on average it occurs as two copies per STAR. The least abundant oligonucleotide, number 166 (AATCGG (SEQ ID NO:342)), occurs on average as a single copy per STAR (thirteen occurrences on eleven STARs); single-copy oligonucleotides occur frequently, especially for the lower-abundance oligos. At the other extreme, oligo 4 (CAGCCC (SEQ ID NO:527)) occurs on average three times in those STARs in which it is found (37 STARs). The most widespread oligonucleotide is number 120 (CCCAGG (SEQ ID NO:296)), which occurs on 58 STARs (on average twice per STAR), and the least widespread oligonucleotide is number 114 (CGTCGC (SEQ ID NO:290)), which occurs on only six STARs (and on average only once per STAR).
Results of dyad frequency analysis are given in Table 7. Seven hundred thirty (730) dyads were found to be over-represented in the set of STAR elements (sig ≧0) compared to the reference sequence. The most significantly over-represented dyad, CCCN{2}CGG (SEQ ID NO:343), occurs 36 times among the 65 STAR elements, but is expected to occur only seven times. It has a significance coefficient of 9.31; in other words, the probability that its over-representation is due to chance is 1/109.31, i.e., less than one in two billion.
Three hundred ninety-seven (397) of the dyads have a significance coefficient greater than 1, and are, therefore, highly over-represented in the STAR elements. Among the over-represented dyads, their observed and expected occurrences, respectively, range from 9 and 1 (for five dyads (numbers 380, 435, 493, 640, and 665)) to 118 and 63 (for number 30 (AGGN{2}GGG (SEQ ID NO:372)), sig=4.44).
The oligonucleotides and dyads found to be over-represented in STAR elements by pattern frequency analysis were tested for their discriminative power by Linear Discriminant Analysis. Discriminant models were trained by step-wise selection of the best combination among the 50 most discriminant oligonucleotide (Table 6) or dyad (Table 7) patterns. The models achieved optimal error rates after incorporation of four (dyad) or five variables. The discriminative variables from oligo analysis are numbers 11, 30, 94, 122, and 160 (Table 6); those from dyad analysis are numbers 73, 194, 419, and 497 (Table 7).
The discriminant models were then used to classify the 65 STAR elements in the training set and their associated random sequences. The model using oligonucleotide variables classifies 46 of the 65 STAR elements as STAR elements (true positives); the dyad model classifies 49 of the STAR elements as true positives. In combination, the models classify 59 of the 65 STAR elements as STAR elements (91%; FIG. 18). The false positive rates (random sequences classified as STARs) were seven for the dyad model, eight for the oligonucleotide model, and 13 for the combined predictions of the two models (20%). The STAR elements of Table 3 that were not classified as STARs by LDA are STAR7, STAR22, STAR35, STAR44, STAR46, and STAR65 (SEQ ID NOS:7, 22, 35, 44, 46 and 65, respectively). These elements display stabilizing anti-repressor activity in functional assays, so the fact that they are not classified as STARs by LDA suggests that they represent another class (or classes) of STAR elements.
The models were then used to classify the 19 candidate STAR elements in the testing set listed in Table 8. The dyad model classifies 12 of these candidate STARs as STAR elements, and the oligonucleotide model classifies 14 as STARs. The combined number of the candidates that are classified as STAR elements is 15 (79%). This is a lower rate of classification than obtained with the training set of 65 STARs; this is expected for two reasons. First, the discriminant models were trained with the 65 STARs of Table 3, and discriminative variables based on this training set may be less well represented in the testing set. Second, the candidate STAR sequences in the testing set have not yet been fully characterized in terms of in vivo function, and may include elements with only weak anti-repression properties.
This analysis demonstrates the power of a statistical approach to bio-informatic classification of STAR elements. The STAR sequences contain a number of dyad and hexameric oligonucleotide patterns that are significantly over-represented in comparison with the human genome as a whole. These patterns may represent binding sites for proteins that confer STAR activity; in any case they form a set of sequence motifs that can be used to recognize STAR element sequences.
Using these patterns to recognize STAR elements by Discriminant Analysis, a high proportion of the elements obtained by the genetic screen of the invention are in fact classified as STARs. This reflects underlying sequence and functional similarities among these elements. An important aspect of the method described here (pattern frequency analysis followed by Discriminant Analysis) is that it can be reiterated; for example, by including the 19 candidate STAR elements of Table 8 with the 66 STAR elements of Table 3 into one training set, an improved discriminant model can be trained. This improved model can then be used to classify other candidate regulatory elements as STARs. Large-scale in vivo screening of genomic sequences using the method of the invention, combined with reiteration of the bio-informatic analysis, will provide a means of discriminating STAR elements that asymptotically approaches 100% recognition and prediction of elements as the genome is screened in its entirety. These stringent and comprehensive predictions of STAR function will ensure that all human STAR elements are recognized, and are available for use in improving transgene expression.
Example 13 Cloning and Characterization of STAR Elements from Arabidopsis thaliana
Transgene silencing occurs in transgenic plants at both the transcriptional and post-transcriptional levels (Meyer, 2000, Vance and Vaucheret, 2001). In either case, the desired result of transgene expression can be compromised by silencing; the low expression and instability of the transgene results in poor expression of desirable traits (e.g., pest resistance) or low yields of recombinant proteins. It also results in poor predictability: the proportion of transgenic plants that express the transgene at biotechnologically useful levels is low, which necessitates laborious and expensive screening of transformed individuals for those with beneficial expression characteristics. This example describes the isolation of STAR elements from the genome of the dicot plant Arabidopsis thaliana for use in preventing transcriptional transgene silencing in transgenic plants. Arabidopsis was chosen for this example because it is a well-studied model organism: it has a compact genome, it is amenable to genetic and recombinant DNA manipulations, and its genome has been sequenced (Bevan et al., 2001, Initiative, 2000, Meinke et al., 1998).
Materials and Methods
Genomic DNA was isolated from Arabidopsis thaliana ecotype Columbia as described (Stam et al., 1998) and partially digested with MboI. The digested DNA was size-fractionated to 0.5-2 kilobase pairs by agarose gel electrophoresis and purification from the gel (QIAquick® Gel Extraction Kit, QIAGEN® 28706), followed by ligation into the pSelect vector (supra). Transfection into the U-2 OS/Tet-Off/LexA-HP1 cell line and selection for zeocin resistance at low doxycycline concentration was performed as described (supra). Plasmids were isolated from zeocin resistant colonies and re-transfected into the U-2 OS/Tet-Off/LexA-HP1 cell line.
Sequencing of Arabidopsis genomic DNA fragments that conferred zeocin resistance upon re-transfection was performed as described (supra). The DNA sequences were compared to the sequence of the Arabidopsis genome by BLAST analysis ((Altschul et al., 1990); URL http://www.ncbi.nlm.nih.gov/blast/Blast).
STAR activity was tested further by measuring mRNA levels for the hygromycin- and zeocin-resistance genes in recombinant host cells by reverse transcription PCR (RT-PCR). Cells of the U-2 OS/Tet-Off/lexA-HP1 cell line were transfected with pSelect plasmids containing Arabidopsis STAR elements, the Drosophila scs element, or containing no insert (supra). These were cultivated on hygromycin for two weeks at high doxycycline concentration, then the doxycycline concentration was lowered to 0.1 ng/ml to induce the lexA-HP1 repressor protein. After ten days, total RNA was isolated by the RNeasy® mini kit (QIAGEN® 74104) as described by the manufacturer. First-strand cDNA synthesis was carried out using the RevertAid™ First Strand cDNA Synthesis kit (MBI Fermentas 1622) using oligo(dT)18 primer as described by the manufacturer. An aliquot of the cDNA was used as the template in a PCR reaction using primers D58 (SEQ ID NO:145) and D80 (SEQ ID NO:148) (for the zeocin marker), and D70 (SEQ ID NO:146) and D71 (SEQ ID NO:147) (for the hygromycin marker), and Taq DNA polymerase (Promega M2661). The reaction conditions were 15-20 cycles of 94° C. for one minute, 54° C. for one minute, and 72° C. for 90 seconds. These conditions result in a linear relationship between input RNA and PCR product DNA. The PCR products were resolved by agarose gel electrophoresis, and the zeocin and hygromycin bands were detected by Southern blotting as described (Sambrook et al., 1989), using PCR products produced as above with purified pSelect plasmid as template. The ratio of the zeocin and hygromycin signals corresponds to the normalized expression level of the zeocin gene.
Results
The library of Arabidopsis genomic DNA in the pSelect vector comprised 69,000 primary clones in E. coli, 80% of which carried inserts. The average insert size was approximately 1000 base pairs; the library, therefore, represents approximately 40% of the Arabidopsis genome.
A portion of this library (representing approximately 16% of the Arabidopsis genome) was transfected into the U-2 OS/Tet-Off/LexA-HP1 cell line. Hygromycin selection was imposed to isolate transfectants, which resulted in 27,000 surviving colonies. These were then subjected to zeocin selection at low doxycycline concentration. Putative STAR-containing plasmids from 56 zeocin-resistant colonies were rescued into E. coli and re-transfected into U-2 OS/Tet-Off/LexA-HP1 cells. Forty-four of these plasmids (79% of the plasmids tested) conferred zeocin resistance on the host cells at low doxycycline concentrations, demonstrating that the plasmids carried STAR elements. This indicates that the pSelect screen in human U-2 OS cells is highly efficient at detection of STAR elements from plant genomic DNA.
The DNA sequences of these 44 candidate STAR elements were determined. Thirty-five of them were identified as single loci in the database of Arabidopsis nuclear genomic sequence (Table 9; SEQ ID NO:85-SEQ ID NO:119). Four others were identified as coming from the chloroplast genome, four were chimeras of DNA fragments from two loci, and one was not found in the Arabidopsis genome database.
The strength of the cloned Arabidopsis STAR elements was tested by assessing their ability to prevent transcriptional repression of the zeocin-resistance gene, using an RT-PCR assay. As a control for RNA input among the samples, the transcript levels of the hygromycin-resistance gene for each STAR transfection were assessed too. This analysis has been performed for 12 of the Arabidopsis STAR elements. The results (FIG. 19) demonstrate that the Arabidopsis STAR elements are superior to the Drosophila scs element (positive control) and the empty vector (“SV40”; negative control) in their ability to protect the zeocin-resistance gene from transcriptional repression. In particular, STAR-A28 (SEQ ID NO:112) and STAR-A30 (SEQ ID NO:114) enable two-fold higher levels of zeocin-resistance gene expression than the scs element (normalized to the internal control of hygromycin-resistance gene mRNA) when the lexA-HP1 repressor is expressed.
These results demonstrate that the method of the invention can be successfully applied to recovery of STAR elements from genomes of other species than human. Its successful application to STAR elements from a plant genome is particularly significant because it demonstrates the wide taxonomic range over which the method of the invention is applicable and because plants are an important target of biotechnological development.
Example 14 STAR Elements Function in CHO Cells
STAR elements function to block the effect of transcriptional repression influences on transgene expression units. Two of the benefits of STAR elements for heterologous protein production are an increased predictability to find high-expressing primary recombinant host cells as well as increased protein production or yield in these cells. Importantly, the disclosed STAR elements are human DNA sequences, isolated in the human U-2 OS osteosarcoma cell line. It is, therefore, an important question whether the human STAR elements are functional in a) cell lines derived from species other than man, and/or in b) human cell lines other than the U-2 OS osteosarcoma cell line. In this example the functionality of STAR 7 (SEQ ID NO:7) in (CHO) Chinese hamster ovary are illustrated.
Material and Methods
The STAR7 (SEQ ID NO:7) element is tested in the ppGIZ-STAR7 vector (FIG. 20). The construction of the pPlug&Play-GFP-ires-Zeo (ppGIZ) vector is described below. Plasmid pGFP (Clontech 6010-1) is modified by insertion of a linker at the BsiWI site to yield pGFP-link. The linker (made by annealing oligonucleotides 5′ GTACGGATATCAGATCTTTAATTAAG 3′ (SEQ ID NO:124) and 5′ GTACCTTAATTAAAGATCTGATATCC 3′ (SEQ ID NO:125)) introduces sites for the PacI, BglII, and EcoRV restriction endonucleases. This creates the multiple cloning site MCSII for insertion of STAR elements. Then primers 5′ ATCAGATCTGGCGCGCCATTTAAATCGTC TCGCGCGTTTCGGTGATGACGG 3′ (SEQ ID NO:126) and 5′ AGGCGGATCCGAATG TATTTAGAAAAATAAACAAATAGGGG 3′ (SEQ ID NO:127) are used to amplify a region of 0.37 kb from pGFP, which is inserted into the BglII site of pIRES (Clontech 6028-1) to yield pIRES-stuf. This introduces sites for the AscI and SwaI restriction endonucleases at MCSI, and acts as a “stuffer fragment” to avoid potential interference between STAR elements and adjacent promoters. pIRES-stuf is digested with BglII and FspI to liberate a DNA fragment composed of the stuffer fragment, the CMV promoter, the IRES element (flanked by multiple cloning sites MCS A and MCS B), and the SV40 polyadenylation signal. This fragment is ligated with the vector backbone of pGFP-link produced by digestion with BamHI and StuI, to yield pIRES-link.
The open reading frames of the zeocin-resistance gene is inserted into the BamHI/NotI sites of MCS B in pIRES-link as follows: the zeocin-resistance ORF is amplified by PCR with primers 5′ GATCGGATCCTTCGAAATGGCCAAGTTGACCAGTGC 3′ (SEQ ID NO:128) and 5′ AGGCGCGGCCGCAATTCTCAGTCCTGCTCCTC 3′ (SEQ ID NO:129) from plasmid pEM7/zeo, digested with BamHI and NotI, and ligated with BamHI/NotI-digested pIRES-link to yield pIRES-link-zeo. The GFP reporter ORF was introduced into pIRES-link-zeo by amplification of phr-GFP-1 with primers 5′ GATCGAATTCTCGCGAATGGTGAGCAAGCAGATCCTGAAG 3′ (SEQ ID NO:130) and 5′ AGGCGAATTCACCGGTGTTTAAACTTACACCCACTCGTGCAGGCTGCCCAGG 3′ (SEQ ID NO:131), and insertion of the EcoRI-digested GFP cassette into the EcoRI site in MCS A of the pIRES-link-zeo plasmid. This created the ppGIZ (for ppGFP-IRES-zeo). STAR7 (SEQ ID NO:7) is cloned into the SalI site (5′) and into the PacI site (3′).
Transfection and Culture of CHO Cells
The Chinese Hamster Ovary cell line CHO-K1 (ATCC CCL-61) is cultured in HAMS-F12 medium+10% Fetal Calf Serum containing 2 mM glutamine, 100 U/ml penicillin, and 100 micrograms/ml streptomycin at 37° C./5% CO2. Cells are transfected with the plasmids using Lipofectamine 2000 (Invitrogen) as described by the manufacturer. Briefly, cells are seeded to culture vessels and grown overnight to 70-90% confluence. Lipofectamine reagent is combined with plasmid DNA at a ratio of 7.5 microliters per 3 microgram (e.g., for a 10 cm Petri dish, 20 micrograms DNA and 120 microliters Lipofectamine) and added after a 30-minute incubation at 25° C. to the cells. After a six-hour incubation, the transfection mixture is replaced with fresh medium, and the transfected cells are incubated further. After overnight cultivation, cells are trypsinized and seeded into fresh petri dishes with fresh medium with zeocin added to a concentration of 100 μg/ml and the cells are cultured further. When individual colonies become visible (approximately ten days after transfection) medium is removed and replaced with fresh medium without zeocin. Individual clones are isolated and transferred to 24-well plates in medium with zeocin. Expression of the GFP reporter gene is assessed approximately three weeks after transfection.
The tested constructs consist of a bicistronic gene with the GFP gene, an IRES and the Zeocin resistance gene under control of the CMV promoter, but either with or without STAR7 (SEQ ID NO:7) element to flank the entire construct (FIG. 20). The constructs are transfected to CHO-K1 cells. Stable colonies are expanded before the GFP signal is determined on a XL-MCL Beckman Coulter flow cytometer. The mean of the GFP signal is taken as measure for the level of GFP expression and this is plotted in FIG. 20.
Results
FIG. 20 shows that flanking a GFP reporter gene that is under the control of the CMV promoter results in a higher number of CHO colonies that express significantly higher levels of GFP protein, as compared to the control without STAR7 (SEQ ID NO:7) element. The STAR7 (SEQ ID NO:7) element, therefore, conveys a higher degree of predictability of transgene expression in CHO cells. The highest GFP expression level in STAR-shielded CHO colonies is also higher than in STAR-less control colonies. In addition, when the tested colonies were further grown for another 30 days without Zeocin in the culture medium, the GFP expression levels in the STAR-shielded colonies remained equally high, whereas the GFP expression levels in the STAR-less colonies dropped to at least below 50% of the original values. It is, therefore, concluded that STAR7 (SEQ ID NO:7) is able to convey higher as well as more stable expression levels to a transgene in CHO cells, this being a cell line derived from another species than man.
Example 15 STAR Elements Function in NSO Cells
STAR elements function to block the effect of transcriptional repression influences on transgene expression units. Two of the benefits of STAR elements for heterologous protein production are an increased predictability to find high-expressing primary recombinant host cells, as well as increased protein production or yield in these cells. Importantly, the disclosed STAR elements are human DNA sequences, isolated in the human U-2 OS osteosarcoma cell line. It is, therefore, an important question whether the human STAR elements are functional in a) cell lines derived from species other than man, and/or in b) human cell lines other than the U-2 OS osteosarcoma cell line. In this example the functionality of STAR 7 (SEQ ID NO:7) in non-secreting mouse myeloma (NSO) cells are illustrated.
Materials and Methods
The tested constructs are the same as described in Example 14. NSO (Non-Secreting mouse myeloma) cells (ECACC 85110503) are suspension cells that are cultured in RPMI 1640 medium+10% Fetal Calf Serum containing 2 mM glutamine, 100 U/ml penicillin, and 100 micrograms/ml streptomycin at 37° C./5% CO2. Cells are transfected with the plasmids using Lipofectamine 2000 (Invitrogen) as described by the manufacturer. Briefly, cells are seeded to culture vessels and grown overnight to 4×105/ml. Lipofectamine reagent is combined with plasmid DNA at a ratio of 3 microliters per microgram DNA (e.g., for a 10 cm Petri dish, 20 micrograms DNA and 60 microliters Lipofectamine) and added after 30 minutes incubation at 25° C. temperature to the cells. After overnight incubation, the transfection mixture is replaced with fresh medium and the transfected cells are incubated further. After another overnight incubation, zeocin is added to a concentration of 100 μg/ml and the cells are cultured and further incubated for three days. Then the cells are seeded in 96-well plates in such dilutions that one well will contain ˜1 cell. After ten days growing colonies are transferred to 24-well plates.
Results
FIG. 21 shows that flanking a GFP reporter gene that is under the control of the CMV promoter results in a higher number of NSO colonies that express significantly higher levels of GFP protein, as compared to the control without STAR7 (SEQ ID NO:7) element. The STAR7 (SEQ ID NO:7) element, therefore, conveys a higher degree of predictability of transgene expression in NSO cells. The highest GFP expression level in STAR-shielded NSO colonies is also higher than in STAR-less control colonies. It is, therefore, concluded that STAR7 (SEQ ID NO:7) is able to convey higher expression levels to a transgene in NSO cells, this being a cell line derived from another species than man.
Example 16 STAR Elements Function in Human 293 Cells
STAR elements function to block the effect of transcriptional repression influences on transgene expression units. Two of the benefits of STAR elements for heterologous protein production are an increased predictability to find high-expressing primary recombinant host cells as well as increased protein production or yield in these cells. Importantly, the disclosed STAR elements are human DNA sequences, isolated in the human U-2 OS osteosarcoma cell line. It is, therefore, an important question whether the human STAR elements are functional in a) cell lines derived from species other than man, and/or in b) human cell lines other than the U-2 OS osteosarcoma cell line. In this example, the functionality of STAR7 (SEQ ID NO:7) in human 293 cells are illustrated.
Materials and Methods
The tested constructs are the same as described in Example 14. The 293 cell line (ATCC CRL-1573) is derived from human embryonal kidney (immortalized by adenovirus 5 transfection) and is cultured in Dulbecco's Modified Eagle Medium+10% Fetal Calf Serum containing 2 mM glutamine, 100 U/ml penicillin, and 100 micrograms/ml streptomycin at 37° C./5% CO2. Cells are transfected with the plasmids using Lipofectamine 2000 (Invitrogen) as described by the manufacturer. Selection and propagation of the 293 colonies are as described in Example 14 for U-2 OS cells.
Results
FIG. 22 shows that flanking a GFP reporter gene that is under the control of the CMV promoter results in a higher number of 293 colonies that express significantly higher levels of GFP protein, as compared to the control without STAR7 (SEQ ID NO:7) element. The STAR7 (SEQ ID NO:7) element, therefore, conveys a higher degree of predictability of transgene expression in 293 cells. The highest GFP expression level in STAR-shielded colonies is also higher than in STAR-less control colonies. It is, therefore, concluded that STAR7 (SEQ ID NO:7) is able to convey higher expression levels to a transgene in 293 cells, this being another human cell line, distinct from the human U-2 OS cell line.
TABLE 1
Biopharmaceutical Proteins, Their Tissue or Cell Type of Origin
Protein Tissue/Somatic Cells Cell Lines (ATCC #)1 Indications
alpha-1 Antitrypsin Liver, leukocytes Hep G2 (HB-8065) Cystic fibrosis, emphysema
alpha-Galactosidase A; -Glucosidase Fibroblasts WI 38 (CCL-75) Fabry disease; Pompe's
disease
Antibodies (monoclonal, Lymphocytes Transfectomas Various therapeutic
single-chain, etc) strategies
Antithrombin III Liver Hep G2 (HB-8065) Thrombophilia
Calcitonin Thyroid (parafollicular cells) TT (CRL-1803) Osteoporosis
Ciliary neurotrophic factor Neural tissue (e.g., astrocytes) HCN-1A (CRL-10442) Motor neuron disease
Epidermal Growth Factor Kidney G-401 (CRL-1441) Wound healing
Erythropoietin Liver, kidney Hep G2 (HB-8065), Anemia
G-401 (CRL-1441)
Factors VII, VIII, IX Endothelial cells HUV-EC-C (CRL-1730) Hemophilia
Famoxin (recombinant gAcrp30) Adipocytes NA2 Obesity
Fibroblast growth factor (basic) Cerebral cortex, hypothalamus HCN-1A (CRL-10442) Wound healing, angiogenesis
Gastric lipase Pancreas BxPC-3 (CRL-1687) Pancreatic insufficiency,
cystic fibrosis
Glucocerebrosidase Macrophages U-937 (CRL-1593.2) Gaucher disease
Granulocyte macrophage-colony T-lymphocytes J.CaM1.6 (CRL-2063) Chemotherapy neutropenia
stimulatin factor
Human growth hormone Pituitary gland HP75 (CRL-2506) Growth retardation, Turner's
(somatotropin) syndrome
Human serum albumin Liver (hepatocytes) Hep G2 (HB-8065) Blood replacement (surgery,
burns)
Insulin Pancreas (Islet beta cells) BxPC-3 (CRL-1687) Diabetes
Interferons alpha Leukocytes WBC264-9C (HB-8902) Cancer, hepatitis C
Interferons beta Fibroblasts WI 38 (CCL-75) Multiple sclerosis
Interleukin-2, -4, -10 T-lymphocytes J.CaM1.6 (CRL-2063) Cancer, rheumatoid arthritis,
hepatitis
Interleukin-18 Monocytes and macrophages U-937 (CRL-1593.2) Cancer, bacterial infections
Interleukin-1 Receptor Antagonist Epithelium HBE4-E6/E7 (CRL-2078) Rheumatoid arthritis
Soluble Tumor Necrosis Factor Placenta, spleen, fibroblasts BeWo (CCL-98) Rheumatoid arthritis,
receptor multiple sclerosis
van Willebrand's factor Endothelial cells HUV-EC-C (CRL-1730) Hemophilia
1These cell lines are offered only as examples of cultured cells corresponding to the tissues and somatic cells; ATCC #: American Type Culture Collection accession number
2NA: Not Available; adipocytes can be differentiated from various other cell types
TABLE 2
Oligonucleotides used for polymerase chain
reactions (PCR primers) or DNA mutagenesis
(SEQ ID NOS:134-176)
SEQ ID NO: Number Sequence
134 C65 AACAAGCTTGATATCAGATCTGCTAGCTTGGT
CGAGCTGATACTTCCC
135 C66 AAACTCGAGCGGCCGCGAATTCGTCGACTTTA
CCACTCCCTATCAGTGATAGAG
136 C67 AAACCGCGGCATGGAAGACGCCAAAAACATAA
AGAAAGG
137 C68 TATGGATCCTAGAATTACACGGCGATCTTTCC
138 C81 AAACCATGGCCGAGTACAAGCCCACGGTGCG
CC
139 C82 AAATCTAGATCAGGCACCGGGCTTGCGGGTCA
TGC
140 C85 CATTTCCCCGAAAAGTGCCACC
141 D30 TCACTGCTAGCGAGTGGTAAACTC
142 D41 GAAGTCGACGAGGCAGGCAGAAGTATGC
143 D42 GAGCCGCGGTTTAGTTCCTCACCTTGTCG
144 D51 TCTGGAAGCTTTGCTGAAGAAAC
145 D58 CCAAGTTGACCAGTGCC
146 D70 TACAAGCCAACCACGGCCT
147 D71 CGGAAGTGCTTGACATTGGG
148 D80 GTTCGTGGACACGACCTCCG
149 D89 GGGCAAGATGTCGTAGTCAGG
150 D90 AGGCCCATGGTCACCTCCATCGCTACTGTG
151 D91 CTAATCACTCACTGTGTAAT
152 D93 AATTACAGGCGCGCC
153 D94 AATTGGCGCGCCTGT
154 D95 TGCTTTGCATACTTCTGCCTGCCTC
155 E12 TAGGGGGGATCCAAATGTTC
156 E13 CCTAAAAGAAGATCTTTAGC
157 E14 AAGTGTTGGATCCACTTTGG
158 E15 TTTGAAGATCTACCAAATGG
159 E16 GTTCGGGATCCACCTGGCCG
160 E17 TAGGCAAGATCTTGGCCCTC
161 E18 CCTCTCTAGGGATCCGACCC
162 E19 CTAGAGAGATCTTCCAGTAT
163 E20 AGAGTTCCGGATCCGCCTGG
164 E21 CCAGGCAGACTCGGAACTCT
165 E22 TGGTGAAACCGGATCCCTAC
166 E23 AGGTCAGGAGATCTAGACCA
167 E25 CCATTTTCGCTTCCTTAGCTCC
168 E42 CGATGTAACCCACTCGTGCACC
169 E57 AGAGATCTAGGATAATTTCG
170 E92 AGGCGCTAGCACGCGTTCTACTCTTTTCCTAC
TCTG
171 E93 GATCAAGCTTACGCGTCTAAAGGCATTTTATA
TAG
172 E94 AGGCGCTAGCACGCGTTCAGAGTTAGTGATCC
AGG
173 E95 GATCAAGCTTACGCGTCAGTAAAGGTTTCGTA
TGG
174 E96 AGGCGCTAGCACGCGTTCTACTCTTTCATTAC
TCTG
175 E97 CGAGGAAGCTGGAGAAGGAGAAGCTG
176 E98 CAAGGGCCGCAGCTTACACATGTTC
TABLE 3
STAR elements of the invention, including genomic location and
length (SEQ ID NOS: 1-66)
STAR SEQ ID NO: Location1 Length
1 1 2q31.1 750
2 2 7p15.2 916
3 3 15q11.2 and 10q22.2 2132
4 4 1p31.1 and 14q24.1 1625
5 5 20q13.32 1571
6 6 2p21 1173
7 7 1q34 2101
8 8 9q32 1839
9 9 10p15.3 1936
10 10 Xp11.3 1167
11 11 2p25.1 1377
12 12 5q35.3 1051
13 13 9q34.3 1291
14 14 22q11.22 732
15 15 1p36.31 1881
16 16 1p21.2 1282
17 17 2q31.1 793
18 18 2q31.3 497
19 19 6p22.1 1840
20 20 8p13.3 780
21 21 6q24.2 620
22 22 2q12.2 1380
23 23 6p22.1 1246
24 24 1q21.2 948
25 25 1q21.3 1067
26 26 1q21.1 540
27 27 1q23.1 1520
28 28 22q11.23 961
29 29 2q13.31 2253
30 30 22q12.3 1851
31 31 9q34.11 and 22q11.21 1165
32 32 21q22.2 771
33 33 21q22.2 1368
34 34 9q34.14 755
35 35 7q22.3 1211
36 36 21q22.2 1712
37 37 22q11.23 1331
38 38 22q11.1 and 22q11.1 ~1000
39 39 22q12.3 2331
40 40 22q11.21 1071
41 41 22q11.21 1144
42 42 22q11.1 735
43 43 14q24.3 1231
44 44 22q11.1 1591
45 45 22q11.21 1991
46 46 22q11.23 1871
47 47 22q11.21 1082
48 48 22q11.22 1242
49 49 Chr 12 random clone, and 1015
3q26.32
50 50 6p21.31 2361
51 51 5q21.3 2289
52 52 7p15.2 1200
53 53 Xp11.3 1431
54 54 4q21.1 981
55 55 15q13.1 501
56 56 includes 3p25.3 741
57 57 4q35.2 1371
58 58 21q11.2 1401
59 59 17 random clone 872
60 60 4p16.1 and 6q27 2068
61 61 7p14.3 and 11q25 1482
62 62 14q24.3 1011
63 63 22q13.3 1421
64 64 17q11.2 1414
65 65 7q21.11 = 28.4 1310
66 66 20q13.33 and 6q14.1 ~2800
1Chromosomal location is determined by BLAST search of DNA sequence data from the STAR elements against the human genome database. The location is given according to standard nomenclature referring to the cytogenetic ideogram of each chromosome; e.g., 1p2.3 is the third cytogenetic sub-band of the second cytogenetic band of the short arm of chromosome 1 (http://www.ncbi.nlm.nih.gov/Class/MLACourse/Genetics/chrombanding.html). F, forward sequencing reaction result; R, reverse sequencing reaction result.
TABLE 4
STAR elements convey stability over time on transgene
expression1
Cell Divisions2 Luciferase Expression3
STAR6 (SEQ ID NO: 6) 42 18,000
plus puromycin 60 23,000
84 20,000
108 16,000
STAR6 (SEQ ID NO: 6) 84 12,000
without puromycin4 108 15,000
144 12,000
1Plasmid pSDH-Tet-STAR6 was transfected into U-2 OS cells, and clones were isolated and cultivated in doxycycline-free medium as described in Example 1. Cells were transferred to fresh culture vessels weekly at a dilution of 1:20.
2The number of cell divisions is based on the estimation that in one week the culture reaches cell confluence, which represents ~6 cell divisions.
3Luciferase was assayed as described in Example 1.
4After 60 cell divisions the cells were transferred to two culture vessels; one was supplied with culture medium that contained puromycin, as for the first 60 cell divisions, and the second was supplied with culture medium lacking antibiotic.
TABLE 5
Human STAR elements and their putative mouse orthologs and
paralogs
NUMBER STAR Human1 Mouse2 Similarity3 SEQ ID NO:
1  1 2q31.1 2D 600 bp 69% 1
2  2 7p15.2 6B3 909 bp 89% 2
3  3a 5q33.3 11B2 248 bp 83% 3
4  3b 10q22.2 14B 1. 363 bp 89% 3
2. 163 bp 86%
5  6 2p21 17E4 437 bp 78% 6
6 12 5q35.3 11b1.3 796 bp 66% 12
7 13 9q34.3 2A3 753 bp 77% 13
8 18 2q31.3 2E1 497 bp 72% 18
9 36 21q22.2 16C4 166 bp 79% 36
10 40 22q11.1 6F1 1. 270 bp 75% 40
2. 309 bp 70%
11 50 6p21.31 17B1 1. 451 bp 72% 50
2. 188 bp 80%
3. 142 bp 64%
12 52 7p15.2 6B3 1. 846 bp 74% 52
2. 195 bp 71%
13 53 Xp11.3 XA2 364 bp 64% 53
14 54 4q21.1 5E3 1. 174 bp 80% 54
2. 240 bp 73%
3. 141 bp 67%
4. 144 bp 68%
15 61a 7p14.3 6B3 188 bp 68% 61
1Cytogenetic location of STAR element in the human genome.
2Cytogenetic location of STAR element ortholog in the mouse genome.
3Length of region(s) displaying high sequence similarity, and percentage similarity. In some cases more than one block of high similarity occurs; in those cases, each block is described separately. Similarity <60% is not considered significant.
The patterns are ranked according to significance coefficient. These were determined using RSA-Tools with the sequence of the human genome as reference. Patterns that comprise the most discriminant variables in Linear Discriminant Analysis are indicated with an asterisk. (SEQ ID NOS:177-342)
TABLE 6
Oligonucleotide patterns (6 base pairs) over-represented in
STAR elements.
Number of
Oligonucleotide Observed Expected Significance matching SEQ ID
Number sequence occurrences occurrences coefficient STARs NO:
1 CCCCAC 107 49 8.76 51 177
2 CAGCGG 36 9 7.75 23 178
3 GGCCCC 74 31 7.21 34 179
4 CAGCCC 103 50 7.18 37 180
5 GCCCCC 70 29 6.97 34 181
6 CGGGGC 40 12 6.95 18 182
7 CCCCGC 43 13 6.79 22 183
8 CGGCAG 35 9 6.64 18 184
9 AGCCCC 83 38 6.54 40 185
10 CCAGGG 107 54 6.52 43 186
11 GGACCC * 58 23 6.04 35 187
12 GCGGAC 20 3 5.94 14 188
13 CCAGCG 34 10 5.9 24 189
14 GCAGCC 92 45 5.84 43 190
15 CCGGCA 28 7 5.61 16 191
16 AGCGGC 27 7 5.45 17 192
17 CAGGGG 86 43 5.09 43 193
18 CCGCCC 43 15 5.02 18 194
19 CCCCCG 35 11 4.91 20 195
20 GCCGCC 34 10 4.88 18 196
21 GCCGGC 22 5 4.7 16 197
22 CGGACC 19 4 4.68 14 198
23 CGCCCC 35 11 4.64 19 199
24 CGCCAG 28 8 4.31 19 200
25 CGCAGC 29 8 4.29 20 201
26 CAGCCG 32 10 4 24 202
27 CCCACG 33 11 3.97 26 203
28 GCTGCC 78 40 3.9 43 204
29 CCCTCC 106 60 3.87 48 205
30 CCCTGC * 92 50 3.83 42 206
31 CACCCC 77 40 3.75 40 207
32 GCGCCA 30 10 3.58 23 208
33 AGGGGC 70 35 3.55 34 209
34 GAGGGC 66 32 3.5 40 210
35 GCGAAC 14 2 3.37 13 211
36 CCGGCG 17 4 3.33 12 212
37 AGCCGG 34 12 3.29 25 213
38 GGAGCC 67 34 3.27 40 214
39 CCCCAG 103 60 3.23 51 215
40 CCGCTC 24 7 3.19 19 216
41 CCCCTC 81 44 3.19 43 217
42 CACCGC 33 12 3.14 22 218
43 CTGCCC 96 55 3.01 42 219
44 GGGCCA 68 35 2.99 39 220
45 CGCTGC 28 9 2.88 22 221
46 CAGCGC 25 8 2.77 19 222
47 CGGCCC 28 10 2.73 19 223
48 CCGCCG 19 5 2.56 9 224
49 CCCCGG 30 11 2.41 17 225
50 AGCCGC 23 7 2.34 17 226
51 GCACCC 55 27 2.31 38 227
52 AGGACC 54 27 2.22 33 228
53 AGGGCG 24 8 2.2 18 229
54 CAGGGC 81 47 2.18 42 230
55 CCCGCC 45 21 2.15 20 231
56 GCCAGC 66 36 2.09 39 232
57 AGCGCC 21 6 2.09 18 233
58 AGGCCC 64 34 2.08 32 234
59 CCCACC 101 62 2.05 54 235
60 CGCTCA 21 6 2.03 17 236
61 AACGCG 9 1 1.96 9 237
62 GCGGCA 21 7 1.92 14 238
63 AGGTCC 49 24 1.87 36 239
64 CCGTCA 19 6 1.78 14 240
65 CAGAGG 107 68 1.77 47 241
66 CCCGAG 33 14 1.77 22 242
67 CCGAGG 36 16 1.76 25 243
68 CGCGGA 11 2 1.75 8 244
69 CCACCC 87 53 1.71 45 245
70 CCTCGC 23 8 1.71 20 246
71 CAAGCC 59 32 1.69 40 247
72 TCCGCA 18 5 1.68 17 248
73 CGCCGC 18 5 1.67 9 249
74 GGGAAC 55 29 1.63 39 250
75 CCAGAG 93 58 1.57 49 251
76 CGTTCC 19 6 1.53 16 252
77 CGAGGA 23 8 1.5 19 253
78 GGGACC 48 24 1.48 31 254
79 CCGCGA 10 2 1.48 8 255
80 CCTGCG 24 9 1.45 17 256
81 CTGCGC 23 8 1.32 14 257
82 GACCCC 47 24 1.31 33 258
83 GCTCCA 66 38 1.25 39 259
84 CGCCAC 33 15 1.19 21 260
85 GCGGGA 23 9 1.17 18 261
86 CTGCGA 18 6 1.15 15 262
87 CTGCTC 80 49 1.14 50 263
88 CAGACG 23 9 1.13 19 264
89 CGAGAG 21 8 1.09 17 265
90 CGGTGC 18 6 1.06 16 266
91 CTCCCC 84 53 1.05 47 267
92 GCGGCC 22 8 1.04 14 268
93 CGGCGC 14 4 1.04 13 269
94 AAGCCC * 60 34 1.03 42 270
95 CCGCAG 24 9 1.03 17 271
96 GCCCAC 59 34 0.95 35 272
97 CACCCA 92 60 0.93 49 273
98 GCGCCC 27 11 0.93 18 274
99 ACCGGC 15 4 0.92 13 275
100 CTCGCA 16 5 0.89 14 276
101 ACGCTC 16 5 0.88 12 277
102 CTGGAC 58 33 0.88 32 278
103 GCCCCA 67 40 0.87 38 279
104 ACCGTC 15 4 0.86 11 280
105 CCCTCG 21 8 0.8 18 281
106 AGCCCG 22 8 0.79 14 282
107 ACCCGA 16 5 0.78 13 283
108 AGCAGC 79 50 0.75 41 284
109 ACCGCG 14 4 0.69 7 285
110 CGAGGC 29 13 0.69 24 286
111 AGCTGC 70 43 0.64 36 287
112 GGGGAC 49 27 0.64 34 288
113 CCGCAA 16 5 0.64 12 289
114 CGTCGC 8 1 0.62 6 290
115 CGTGAC 17 6 0.57 15 291
116 CGCCCA 33 16 0.56 22 292
117 CTCTGC 97 65 0.54 47 293
118 AGCGGG 21 8 0.52 17 294
119 ACCGCT 15 5 0.5 11 295
120 CCCAGG 133 95 0.49 58 296
121 CCCTCA 71 45 0.49 39 297
122 CCCCCA * 77 49 0.49 42 298
123 GGCGAA 16 5 0.48 14 299
124 CGGCTC 29 13 0.47 19 300
125 CTCGCC 20 8 0.46 17 301
126 CGGAGA 20 8 0.45 14 302
127 TCCCCA 95 64 0.43 52 303
128 GACACC 44 24 0.42 33 304
129 CTCCGA 17 6 0.42 13 305
130 CTCGTC 17 6 0.42 14 306
131 CGACCA 13 4 0.39 11 307
132 ATGACG 17 6 0.37 12 308
133 CCATCG 17 6 0.37 13 309
134 AGGGGA 78 51 0.36 44 310
135 GCTGCA 77 50 0.35 43 311
136 ACCCCA 76 49 0.33 40 312
137 CGGAGC 21 9 0.33 16 313
138 CCTCCG 28 13 0.32 19 314
139 CGGGAC 16 6 0.3 10 315
140 CCTGGA 88 59 0.3 45 316
141 AGGCGA 18 7 0.29 17 317
142 ACCCCT 54 32 0.28 36 318
143 GCTCCC 56 34 0.27 36 319
144 CGTCAC 16 6 0.27 15 320
145 AGCGCA 16 6 0.26 11 321
146 GAAGCC 62 38 0.25 39 322
147 GAGGCC 79 52 0.22 42 323
148 ACCCTC 54 32 0.22 33 324
149 CCCGGC 37 20 0.21 21 325
150 CGAGAA 20 8 0.2 17 326
151 CCACCG 29 14 0.18 20 327
152 ACTTCG 16 6 0.17 14 328
153 GATGAC 48 28 0.17 35 329
154 ACGAGG 23 10 0.16 18 330
155 CCGGAG 20 8 0.15 18 331
156 ACCCAC 60 37 0.12 41 332
157 CTGGGC 105 74 0.11 50 333
158 CCACGG 23 10 0.09 19 334
159 CGGTCC 13 4 0.09 12 335
160 AGCACC * 54 33 0.09 40 336
161 ACACCC 53 32 0.08 38 337
162 AGGGCC 54 33 0.08 30 338
163 CGCGAA 6 1 0.02 6 339
164 GAGCCC 58 36 0.02 36 340
165 CTGAGC 71 46 0.02 45 341
166 AATCGG 13 4 0.02 11 342
The patterns are ranked according to significance coefficient. These were determined using RSA-Tools with the random sequence from the human genome as reference. Patterns that comprise the most discriminant variables in Linear Discriminant Analysis are indicated with an asterisk. (SEQ ID NOS:343-1072)
TABLE 7
Dyad patterns over-represented in STAR elements.
Observed Expected Significance SEQ ID
Number Dyad sequence occurrences occurrences coefficient NO:
1 CCCN{2}CGG 36 7 9.31 343
2 CCGN{6}CCC 40 10 7.3 344
3 CAGN{0}CGG 36 8 7.13 345
4 CGCN{15}CCC 34 8 6.88 346
5 CGGN{9}GCC 33 7 6.82 347
6 CCCN{9}CGC 35 8 6.72 348
7 CCCN{1}GCG 34 8 6.64 349
8 CCCN{0}CAC 103 48 6.61 350
9 AGCN{16}CCG 29 6 5.96 351
10 CCCN{4}CGC 34 8 5.8 352
11 CGCN{13}GGA 26 5 5.77 353
12 GCGN{16}CCC 30 7 5.74 354
13 CGCN{5}GCA 25 5 5.49 355
14 CCCN{14}CCC 101 49 5.43 356
15 CTGN{4}CGC 34 9 5.41 357
16 CCAN{12}GCG 28 6 5.37 358
17 CGGN{11}CAG 36 10 5.25 359
18 CCCN{5}GCC 75 33 4.87 360
19 GCCN{0}CCC 64 26 4.81 361
20 CGCN{4}GAC 19 3 4.78 362
21 CGGN{0}CAG 33 9 4.76 363
22 CCCN{3}CGC 32 8 4.67 364
23 CGCN{1}GAC 20 3 4.58 365
24 GCGN{2}GCC 29 7 4.54 366
25 CCCN{4}GCC 76 34 4.53 367
26 CCCN{1}CCC 103 52 4.53 368
27 CCGN{13}CAG 33 9 4.5 369
28 GCCN{4}GGA 64 27 4.48 370
29 CCGN{3}GGA 26 6 4.46 371
30 AGGN{2}GGG 118 63 4.44 372
31 CACN{5}GCG 22 4 4.42 373
32 CGCN{17}CCA 27 6 4.39 374
33 CCCN{9}GGC 69 30 4.38 375
34 CCTN{5}GCG 28 7 4.37 376
35 GCGN{0}GAC 19 3 4.32 377
36 GCCN{0}GGC 40 7 4.28 378
37 GCGN{2}CCC 26 6 4.27 379
38 CCGN{11}CCC 32 9 4.17 380
39 CCCN{8}TCG 23 5 4.12 381
40 CCGN{17}GCC 30 8 4.12 382
41 GGGN{5}GGA 101 52 4.11 383
42 GGCN{6}GGA 71 32 4.1 384
43 CCAN{4}CCC 96 48 4.1 385
44 CCTN{14}CCG 32 9 4.09 386
45 GACN{12}GGC 45 16 4.07 387
46 CGCN{13}CCC 30 8 4.04 388
47 CAGN{16}CCC 92 46 4.02 389
48 AGCN{10}GGG 75 35 3.94 390
49 CGGN{13}GGC 30 8 3.93 391
50 CGGN{1}GCC 30 8 3.92 392
51 AGCN{0}GGC 26 6 3.9 393
52 CCCN{16}GGC 64 28 3.89 394
53 GCTN{19}CCC 67 29 3.87 395
54 CCCN{16}GGG 88 31 3.81 396
55 CCCN{9}CGG 30 8 3.77 397
56 CCCN{10}CGG 30 8 3.76 398
57 CCAN{0}GCG 32 9 3.75 399
58 GCCN{17}CGC 26 6 3.74 400
59 CCTN{6}CGC 27 7 3.73 401
60 GGAN{1}CCC 63 27 3.71 402
61 CGCN{18}CAC 24 5 3.7 403
62 CGCN{20}CCG 21 4 3.69 404
63 CCGN{0}GCA 26 6 3.69 405
64 CGCN{20}CCC 28 7 3.69 406
65 AGCN{15}CCC 67 30 3.65 407
66 CCTN{7}GGC 69 31 3.63 408
67 GCCN{5}CGC 32 9 3.61 409
68 GCCN{14}CGC 28 7 3.59 410
69 CAGN{11}CCC 89 45 3.58 411
70 GGGN{16}GAC 53 21 3.57 412
71 CCCN{15}GCG 25 6 3.57 413
72 CCCN{0}CGC 37 12 3.54 414
73 CCCN{16}AGC * 67 30 3.54 415
74 AGGN{9}GGG 96 50 3.52 416
75 CGCN{12}CTC 28 7 3.46 417
76 CACN{8}CGC 23 5 3.43 418
77 CCAN{7}CCG 31 9 3.42 419
78 CGGN{1}GCA 25 6 3.41 420
79 CGCN{14}CCC 29 8 3.4 421
80 AGCN{0}CCC 76 36 3.4 422
81 CGCN{13}GTC 18 3 3.37 423
82 GCGN{3}GCA 26 7 3.35 424
83 CGGN{0}GGC 34 11 3.35 425
84 GCCN{14}CCC 68 31 3.33 426
85 ACCN{7}CGC 21 4 3.32 427
86 AGGN{7}CGG 33 10 3.31 428
87 CCCN{16}CGA 22 5 3.3 429
88 CGCN{6}CAG 31 9 3.29 430
89 CAGN{11}GCG 29 8 3.29 431
90 CCGN{12}CCG 19 4 3.26 432
91 CGCN{18}CAG 27 7 3.24 433
92 CAGN{1}GGG 80 39 3.21 434
93 CGCN{0}CCC 32 10 3.2 435
94 GCGN{18}GCC 26 7 3.18 436
95 CGGN{15}GGC 27 7 3.15 437
96 CCCN{15}AGG 72 34 3.14 438
97 AGGN{20}GCG 26 7 3.14 439
98 CGGN{5}CTC 26 7 3.13 440
99 TCCN{17}CGA 23 5 3.12 441
100 GCGN{4}CCC 30 9 3.08 442
101 CCCN{2}CGC 30 9 3.07 443
102 CGTN{3}CAG 28 8 3.06 444
103 CCGN{13}GAG 27 7 3.05 445
104 CTCN{6}CGC 28 8 3.04 446
105 CGCN{4}GAG 21 5 3.03 447
106 GCGN{5}GGA 24 6 3.03 448
107 CCGN{1}CAG 27 7 3.01 449
108 CGCN{11}CCG 18 3 2.99 450
109 GCGN{19}CCC 26 7 2.98 451
110 CGCN{18}GAA 21 5 2.98 452
111 GGGN{19}GGA 78 39 2.95 453
112 CCAN{1}CGG 24 6 2.94 454
113 CCCN{7}GCG 25 6 2.94 455
114 AGGN{10}CCC 84 43 2.92 456
115 CCAN{0}GGG 97 52 2.88 457
116 CAGN{10}CCC 82 41 2.87 458
117 CCGN{18}CCG 19 4 2.86 459
118 CCGN{18}GGC 26 7 2.85 460
119 CCCN{2}GCG 24 6 2.84 461
120 CGCN{1}GGC 25 7 2.83 462
121 CCGN{5}GAC 19 4 2.81 463
122 GGAN{0}CCC 52 22 2.8 464
123 CCCN{1}CCG 29 9 2.78 465
124 CCCN{15}ACG 23 6 2.75 466
125 AGCN{8}CCC 66 31 2.73 467
126 CCCN{3}GGC 60 27 2.71 468
127 AGGN{9}CGG 31 10 2.7 469
128 CCCN{14}CGC 27 8 2.7 470
129 CCGN{0}CCG 19 4 2.7 471
130 CGCN{8}AGC 23 6 2.69 472
131 CGCN{19}ACC 21 5 2.68 473
132 GCGN{17}GAC 17 3 2.66 474
133 AGCN{1}GCG 24 6 2.63 475
134 CCGN{11}GGC 31 10 2.63 476
135 CGGN{4}AGA 26 7 2.63 477
136 CGCN{14}CCG 17 3 2.62 478
137 CCTN{20}GCG 24 6 2.62 479
138 CCAN{10}CGC 26 7 2.61 480
139 CCCN{20}CAC 69 33 2.6 481
140 CCGN{11}GCC 27 8 2.6 482
141 CGCN{18}CCC 26 7 2.59 483
142 CGGN{15}CGC 16 3 2.57 484
143 CGCN{16}GCC 24 6 2.55 485
144 CGCN{20}GGC 23 6 2.54 486
145 CGCN{19}CCG 18 4 2.52 487
146 CGGN{10}CCA 28 8 2.51 488
147 CGCN{17}CCC 26 7 2.51 489
148 CGCN{11}ACA 23 6 2.51 490
149 CGGN{0}ACC 17 3 2.5 491
150 GCGN{10}GCC 24 6 2.49 492
151 GCGN{8}GAC 17 3 2.49 493
152 CCCN{15}GGG 84 32 2.44 494
153 CGGN{16}GGC 27 8 2.44 495
154 CGCN{16}CCA 23 6 2.42 496
155 GCCN{3}CCC 73 36 2.4 497
156 CAGN{4}GGG 94 51 2.4 498
157 CCCN{6}GCG 23 6 2.38 499
158 CCGN{16}CGC 17 3 2.38 500
159 CCCN{17}GCA 61 28 2.37 501
160 CGCN{13}TCC 24 6 2.37 502
161 GCCN{1}CGC 29 9 2.36 503
162 CCGN{19}GAG 26 7 2.35 504
163 GGGN{10}GGA 89 48 2.35 505
164 CAGN{5}CCG 32 11 2.35 506
165 CGCN{3}AGA 19 4 2.32 507
166 GCCN{0}GCC 29 9 2.32 508
167 CCCN{8}GGC 61 28 2.31 509
168 CCTN{6}GCG 22 6 2.29 510
169 GACN{6}CCC 48 20 2.29 511
170 CGGN{1}CCC 26 8 2.27 512
171 CCCN{15}CCG 30 10 2.27 513
172 CAGN{9}CCC 84 44 2.26 514
173 CGGN{10}GGC 27 8 2.26 515
174 CGAN{10}ACG 10 1 2.26 516
175 GCGN{3}TCC 21 5 2.26 517
176 CCCN{3}GCC 75 38 2.24 518
177 GCGN{1}ACC 17 3 2.24 519
178 CCGN{9}AGG 27 8 2.23 520
179 CGCN{16}CAG 26 8 2.23 521
180 GGCN{0}CCC 62 29 2.22 522
181 AGGN{12}CCG 26 8 2.19 523
182 CCGN{0}GCG 16 3 2.19 524
183 CCGN{2}GCC 30 10 2.18 525
184 CCGN{11}GTC 19 4 2.17 526
185 CAGN{0}CCC 88 47 2.17 527
186 CCCN{5}CCG 32 11 2.17 528
187 GCCN{20}CCC 66 32 2.15 529
188 GACN{2}CGC 18 4 2.14 530
189 CGCN{6}CAC 23 6 2.13 531
190 AGGN{14}GCG 25 7 2.1 532
191 GACN{5}CGC 17 3 2.1 533
192 CCTN{19}CCG 29 9 2.1 534
193 CCGN{12}GGA 24 7 2.08 535
194 GGCN{9}GAC * 44 18 2.08 536
195 AGGN{10}GGG 94 52 2.07 537
196 CCGN{10}GAG 25 7 2.07 538
197 CGCN{6}GGA 20 5 2.06 539
198 CGCN{7}AGC 23 6 2.04 540
199 CCAN{13}CGG 26 8 2.03 541
200 CGGN{6}GGA 25 7 2.03 542
201 CGCN{19}GCC 24 7 2.03 543
202 CCAN{12}CGC 24 7 2.02 544
203 CGGN{1}GGC 41 16 2.02 545
204 GCGN{3}CCA 25 7 2.01 546
205 AGGN{1}CGC 21 5 2 547
206 CTCN{5}CGC 24 7 1.98 548
207 CCCN{0}ACG 30 10 1.97 549
208 CAGN{17}CCG 29 9 1.96 550
209 GGCN{4}CCC 62 30 1.96 551
210 AGGN{8}GCG 26 8 1.96 552
211 CTGN{1}CCC 88 48 1.94 553
212 CCCN{16}CAG 85 46 1.94 554
213 CGCN{9}GAC 16 3 1.93 555
214 CAGN{6}CCG 29 9 1.92 556
215 CGTN{12}CGC 11 1 1.92 557
216 CTCN{7}GCC 69 35 1.92 558
217 CGCN{19}TCC 22 6 1.92 559
218 CCCN{7}GCC 67 33 1.91 560
219 CAGN{13}CGG 30 10 1.9 561
220 CGCN{1}GCC 27 8 1.9 562
221 CGCN{17}CCG 17 4 1.89 563
222 AGGN{4}CCC 63 31 1.89 564
223 AGCN{10}CGC 21 5 1.89 565
224 CCCN{11}CGG 30 10 1.88 566
225 CCCN{8}GCC 75 39 1.86 567
226 CCGN{1}CGG 22 3 1.86 568
227 CCCN{1}ACC 71 36 1.85 569
228 CGCN{0}CAG 25 7 1.85 570
229 CCGN{19}TGC 23 6 1.82 571
230 GCGN{4}CGA 12 2 1.82 572
231 CCGN{19}GCC 30 10 1.82 573
232 CCAN{10}CCC 85 46 1.81 574
233 CAGN{13}GGG 91 51 1.81 575
234 AGCN{18}CGG 23 6 1.81 576
235 CGAN{8}CGC 11 1 1.81 577
236 AGCN{4}CCC 63 31 1.8 578
237 GGAN{6}CCC 61 30 1.8 579
238 CGGN{13}AAG 23 6 1.8 580
239 ACCN{11}CGC 19 5 1.79 581
240 CCGN{12}CAG 28 9 1.78 582
241 CCCN{12}GGG 76 29 1.77 583
242 CACN{17}ACG 22 6 1.76 584
243 CAGN{18}CCC 82 44 1.76 585
244 CGTN{10}GTC 19 5 1.75 586
245 CCCN{13}GCG 23 6 1.75 587
246 GCAN{1}CGC 20 5 1.73 588
247 AGAN{4}CCG 24 7 1.73 589
248 GCGN{10}AGC 22 6 1.72 590
249 CGCN{0}GGA 12 2 1.72 591
250 CGGN{4}GAC 17 4 1.69 592
251 CCCN{12}CGC 26 8 1.68 593
252 GCCN{15}CCC 65 33 1.68 594
253 GCGN{6}TCC 20 5 1.66 595
254 CGGN{3}CAG 33 12 1.65 596
255 CCCN{3}CCA 88 49 1.65 597
256 AGCN{3}CCC 59 28 1.65 598
257 GGGN{16}GCA 65 33 1.65 599
258 AGGN{8}CCG 28 9 1.64 600
259 CCCN{0}CCG 29 10 1.64 601
260 GCGN{5}GAC 16 3 1.64 602
261 CCCN{9}ACC 60 29 1.64 603
262 CTGN{5}CGC 25 8 1.64 604
263 CGCN{14}CTC 23 7 1.64 605
264 CGGN{14}GCA 23 7 1.63 606
265 CCGN{8}GCC 26 8 1.62 607
266 CCGN{7}CAC 23 7 1.62 608
267 AGCN{8}GCG 21 6 1.61 609
268 CGGN{16}GGA 29 10 1.61 610
269 CCAN{12}CCG 26 8 1.61 611
270 CGGN{2}CCC 26 8 1.6 612
271 CCAN{13}GGG 71 37 1.6 613
272 CGGN{15}GCA 21 6 1.6 614
273 CGCN{9}GCA 20 5 1.58 615
274 CGGN{19}CCA 26 8 1.58 616
275 GGGN{15}CGA 20 5 1.57 617
276 CCCN{10}CGC 26 8 1.57 618
277 CTCN{14}CGC 26 8 1.55 619
278 CACN{11}GCG 20 5 1.55 620
279 CCGN{2}GGC 24 7 1.55 621
280 CTGN{18}CCC 85 47 1.54 622
281 GGGN{13}CAC 58 28 1.54 623
282 CCTN{15}GGC 62 31 1.54 624
283 CCCN{20}CGA 20 5 1.54 625
284 CCCN{8}CGA 20 5 1.53 626
285 GAGN{7}CCC 61 30 1.53 627
286 CGCN{2}CCG 22 6 1.53 628
287 CCCN{0}TCC 98 57 1.52 629
288 AGCN{0}GCC 21 6 1.52 630
289 CCCN{2}TCC 82 45 1.52 631
290 CCGN{5}CCC 30 10 1.52 632
291 CGCN{13}CGC 16 3 1.51 633
292 CCCN{1}CGC 28 9 1.51 634
293 GCCN{16}GCA 53 25 1.51 635
294 CCCN{16}CCA 84 46 1.5 636
295 CCGN{13}CGC 19 5 1.5 637
296 CCGN{17}CAG 28 9 1.49 638
297 CGGN{18}GGC 26 8 1.49 639
298 CCGN{14}AGG 23 7 1.49 640
299 CCCN{5}CGG 26 8 1.49 641
300 CCCN{6}GGA 58 28 1.49 642
301 ACGN{2}CCC 20 5 1.49 643
302 CCAN{9}CCG 27 9 1.48 644
303 CCCN{19}CCA 78 42 1.48 645
304 CAGN{0}GGG 77 41 1.48 646
305 AGCN{1}CCC 58 28 1.47 647
306 GCGN{7}TCC 27 9 1.46 648
307 ACGN{18}CCA 25 8 1.46 649
308 GCTN{14}CCC 61 30 1.46 650
309 GCGN{14}CCC 23 7 1.46 651
310 GCGN{19}AGC 20 5 1.45 652
311 CCGN{8}CAG 29 10 1.45 653
312 GCGN{6}GCC 22 6 1.45 654
313 GCGN{10}GCA 20 5 1.44 655
314 CCTN{7}GCC 69 36 1.44 656
315 GCCN{13}GCC 54 26 1.42 657
316 CCCN{14}GCC 63 32 1.42 658
317 CCCN{15}CGG 26 8 1.42 659
318 CCAN{13}CGC 23 7 1.42 660
319 AGCN{11}GGG 67 35 1.41 661
320 GGAN{0}GCC 64 32 1.4 662
321 GCCN{3}TCC 61 30 1.4 663
322 CCTN{5}GCC 69 36 1.39 664
323 CGGN{18}CCC 25 8 1.39 665
324 CCTN{3}GGC 59 29 1.38 666
325 CCGN{0}CTC 22 6 1.38 667
326 AGCN{17}GCG 19 5 1.37 668
327 ACGN{14}GGG 20 5 1.37 669
328 CGAN{12}GGC 19 5 1.37 670
329 CCCN{20}CGC 24 7 1.37 671
330 ACGN{12}CTG 24 7 1.36 672
331 CCGN{0}CCC 36 14 1.36 673
332 CCGN{10}GGA 23 7 1.36 674
333 CCCN{3}GCG 21 6 1.36 675
334 GCGN{14}CGC 22 3 1.35 676
335 CCGN{8}CGC 16 4 1.35 677
336 CGCN{10}ACA 22 6 1.34 678
337 CCCN{19}CCG 28 10 1.33 679
338 CACN{14}CGC 20 5 1.32 680
339 GACN{3}GGC 46 21 1.32 681
340 GAAN{7}CGC 19 5 1.32 682
341 CGCN{16}GGC 21 6 1.31 683
342 GGCN{9}CCC 64 33 1.31 684
343 CCCN{9}GCC 64 33 1.31 685
344 CGCN{0}TGC 26 9 1.3 686
345 CCTN{8}GGC 67 35 1.3 687
346 CCAN{8}CCC 82 46 1.29 688
347 GACN{2}CCC 42 18 1.28 689
348 GGCN{1}CCC 54 26 1.27 690
349 CGCN{0}AGC 24 7 1.26 691
350 AGGN{4}GCG 28 10 1.26 692
351 CGGN{6}TCC 22 6 1.25 693
352 ACGN{19}GGC 20 5 1.25 694
353 CCCN{8}ACG 21 6 1.24 695
354 CCCN{18}GCC 62 31 1.24 696
355 GCCN{2}CGA 19 5 1.24 697
356 CCCN{8}GCG 28 10 1.23 698
357 CCCN{0}CTC 76 41 1.23 699
358 GCCN{11}CGC 27 9 1.22 700
359 AGCN{9}CCC 59 29 1.22 701
360 GCTN{0}GCC 71 38 1.21 702
361 CGCN{3}CCC 26 9 1.21 703
362 CCCN{2}CCC 117 72 1.19 704
363 GCCN{9}CGC 23 7 1.19 705
364 GCAN{19}CGC 19 5 1.19 706
365 CAGN{4}CGG 32 12 1.18 707
366 CAGN{2}GGG 80 44 1.17 708
367 GCCN{16}CCC 67 35 1.16 709
368 GAGN{5}CCC 60 30 1.16 710
369 CCTN{16}TCG 20 6 1.16 711
370 CCCN{2}GGC 62 32 1.15 712
371 GCGN{13}GGA 24 8 1.15 713
372 GCCN{17}GGC 66 25 1.15 714
373 CCCN{14}GGC 58 29 1.14 715
374 AGGN{3}CCG 31 12 1.14 716
375 CACN{0}CGC 32 12 1.14 717
376 CGGN{18}CAG 28 10 1.14 718
377 AGCN{1}GCC 57 28 1.13 719
378 CGCN{18}GGC 23 7 1.13 720
379 CCCN{5}AGG 64 33 1.11 721
380 AACN{0}GCG 9 1 1.11 722
381 CCCN{10}CCA 88 50 1.09 723
382 CGCN{13}GAG 20 6 1.09 724
383 CGCN{7}GCC 25 8 1.08 725
384 CCCN{9}CCG 28 10 1.07 726
385 CGCN{16}CCC 24 8 1.05 727
386 GAAN{13}CGC 18 5 1.05 728
387 GGCN{3}CCC 49 23 1.03 729
388 TCCN{11}CCA 87 50 1.03 730
389 CACN{0}CCC 70 38 1.02 731
390 CGCN{16}CCG 15 3 1.02 732
391 CGGN{15}AGC 21 6 1.02 733
392 CCCN{12}GCG 21 6 1.02 734
393 CCCN{9}GAG 59 30 1.01 735
394 CCGN{20}TCC 24 8 1.01 736
395 CGCN{0}CGC 17 4 1.01 737
396 ATGN{7}CGG 20 6 1 738
397 GGGN{20}GCA 59 30 1 739
398 CGGN{4}GGC 26 9 0.99 740
399 CGGN{16}AGC 22 7 0.99 741
400 CGGN{5}GGC 25 8 0.99 742
401 GCGN{0}GGA 25 8 0.98 743
402 GGCN{20}CAC 52 25 0.98 744
403 CCCN{9}CCC 97 58 0.97 745
404 ACCN{17}GGC 44 20 0.97 746
405 CCCN{6}CGA 18 5 0.96 747
406 AAGN{10}CGG 26 9 0.96 748
407 CGCN{17}CAC 21 6 0.95 749
408 CCCN{16}CGG 25 8 0.94 750
409 GACN{18}GGC 39 17 0.94 751
410 GGGN{15}GAC 47 22 0.92 752
411 GCCN{4}TCC 66 35 0.92 753
412 GGCN{15}CCC 56 28 0.92 754
413 CAGN{12}CGC 24 8 0.92 755
414 CCAN{3}GCG 22 7 0.91 756
415 CCGN{16}GAG 22 7 0.9 757
416 AGCN{2}CGC 24 8 0.89 758
417 GAGN{4}CCC 54 27 0.89 759
418 AGGN{3}CGC 23 7 0.88 760
419 CACN{13}AGG * 67 36 0.88 761
420 CCCN{4}CAG 88 51 0.88 762
421 CCCN{2}GAA 63 33 0.87 763
422 CGCN{19}GAG 21 6 0.87 764
423 ACGN{18}GGG 21 6 0.87 765
424 CCCN{4}GGC 62 32 0.87 766
425 CGGN{9}GAG 28 10 0.86 767
426 CCCN{3}GGG 66 26 0.86 768
427 GAGN{4}GGC 66 35 0.85 769
428 CGCN{5}GAG 18 5 0.84 770
429 CCGN{20}AGG 24 8 0.84 771
430 CCCN{15}CCC 88 51 0.83 772
431 AGGN{17}CCG 25 8 0.82 773
432 AGGN{6}GGG 89 52 0.82 774
433 GGCN{20}CCC 57 29 0.82 775
434 GCAN{17}CGC 19 5 0.82 776
435 CGAN{11}ACG 9 1 0.81 777
436 CGCN{2}GGA 19 5 0.81 778
437 CTGN{5}CCC 79 45 0.8 779
438 TCCN{20}CCA 77 43 0.8 780
439 CCAN{2}GGG 59 30 0.8 781
440 CCGN{15}GCG 14 3 0.8 782
441 CCAN{5}GGG 69 38 0.79 783
442 CGGN{1}TGC 24 8 0.79 784
443 CCCN{14}GCG 21 6 0.79 785
444 CAGN{0}CCG 27 10 0.79 786
445 GCCN{9}TCC 60 31 0.78 787
446 AGGN{20}CGC 22 7 0.78 788
447 CCCN{6}GAC 42 19 0.77 789
448 CGGN{11}CCA 23 7 0.76 790
449 GGGN{14}CAC 57 29 0.75 791
450 GCAN{15}CGC 19 5 0.74 792
451 CGCN{2}ACA 20 6 0.74 793
452 ACCN{9}CCC 57 29 0.73 794
453 GCGN{9}CGC 20 3 0.73 795
454 CAGN{15}GCG 23 7 0.73 796
455 CCCN{18}GTC 45 21 0.72 797
456 GCGN{3}CCC 24 8 0.72 798
457 CGGN{11}GCC 23 8 0.72 799
458 CCCN{1}CGG 24 8 0.71 800
459 GCCN{4}CCA 70 38 0.71 801
460 CCCN{4}CCG 30 12 0.7 802
461 CGTN{2}GCA 21 6 0.7 803
462 AGCN{7}TCG 18 5 0.69 804
463 CCGN{15}GAA 20 6 0.69 805
464 ACCN{5}CCC 62 33 0.69 806
465 CGCN{14}GAG 19 5 0.68 807
466 CCCN{7}CGC 30 12 0.68 808
467 GAGN{12}CGC 21 6 0.68 809
468 GGCN{17}CCC 58 30 0.67 810
469 ACGN{11}CTC 21 7 0.65 811
470 ACAN{9}CGG 24 8 0.65 812
471 CTGN{7}CCC 82 47 0.65 813
472 CCCN{2}GCC 72 40 0.65 814
473 CGGN{2}GCA 24 8 0.64 815
474 CCCN{0}TGC 83 48 0.64 816
475 CGCN{7}ACC 18 5 0.63 817
476 GCAN{2}GCC 54 27 0.63 818
477 GCGN{8}CCA 20 6 0.63 819
478 AGCN{0}CGC 22 7 0.63 820
479 GCGN{2}GCA 18 5 0.63 821
480 CCGN{2}GTC 18 5 0.62 822
481 CCGN{3}ACA 21 7 0.62 823
482 ACGN{13}TGG 21 7 0.62 824
483 CCAN{8}CGC 23 8 0.62 825
484 CCGN{9}GGC 23 8 0.61 826
485 CCAN{5}CCG 25 9 0.61 827
486 AGGN{3}GGG 97 59 0.61 828
487 CAGN{2}GGC 78 45 0.61 829
488 CCCN{8}CAG 81 47 0.61 830
489 AGCN{5}CAG 80 46 0.6 831
490 CGGN{16}GCC 22 7 0.6 832
491 GCGN{15}CCC 23 8 0.6 833
492 CCCN{11}GCC 59 31 0.59 834
493 CGAN{2}ACG 9 1 0.59 835
494 CGGN{4}GCC 22 7 0.59 836
495 CACN{6}CGC 19 6 0.59 837
496 CGGN{5}ACG 11 2 0.59 838
497 CTGN{4}GCC * 66 36 0.59 839
498 GGGN{18}CGA 18 5 0.59 840
499 CCTN{8}CGC 22 7 0.59 841
500 GCCN{4}CCC 67 37 0.58 842
501 CGGN{10}GCC 22 7 0.58 843
502 GCCN{5}GGA 54 27 0.57 844
503 ACCN{7}GCG 15 4 0.57 845
504 CCCN{8}CGC 24 8 0.57 846
505 CAGN{5}CCC 77 44 0.56 847
506 CACN{14}GGA 63 34 0.56 848
507 CCCN{1}GCC 94 57 0.55 849
508 CCCN{5}AGC 67 37 0.55 850
509 GGCN{5}GGA 59 31 0.55 851
510 CGAN{17}GAG 19 6 0.55 852
511 CGCN{7}ACA 18 5 0.54 853
512 CCAN{13}CCC 87 52 0.54 854
513 CGGN{20}GGC 24 8 0.54 855
514 CCCN{17}GCC 58 30 0.53 856
515 CCTN{10}CCG 30 12 0.53 857
516 CCCN{8}CCG 27 10 0.53 858
517 CGCN{3}GAG 18 5 0.52 859
518 CGCN{7}AAG 17 5 0.51 860
519 CGGN{11}GGA 23 8 0.51 861
520 CCGN{15}CCG 15 4 0.51 862
521 CCCN{3}GCA 57 30 0.51 863
522 CGGN{2}CAG 24 8 0.5 864
523 AGGN{2}CCG 24 8 0.5 865
524 CCCN{4}CAC 69 38 0.5 866
525 GGAN{19}CCC 56 29 0.49 867
526 CCCN{8}CAC 68 38 0.49 868
527 ACCN{6}CCG 18 5 0.49 869
528 CCCN{6}GGC 54 28 0.49 870
529 CCCN{6}CCG 29 11 0.48 871
530 CGCN{14}GCC 26 9 0.47 872
531 CCGN{5}TCC 25 9 0.46 873
532 GCCN{6}GCC 55 28 0.46 874
533 CGGN{7}GGA 24 8 0.45 875
534 GGGN{6}GGA 87 52 0.44 876
535 GCCN{12}TCC 60 32 0.44 877
536 AGTN{16}CCG 17 5 0.44 878
537 GGCN{19}GCC 68 29 0.44 879
538 CCGN{3}CCG 22 7 0.44 880
539 CCCN{8}ACC 58 31 0.44 881
540 CAGN{15}GCC 77 44 0.44 882
541 CCCN{17}CGG 24 8 0.44 883
542 GCGN{1}CCA 22 7 0.44 884
543 CCCN{14}CAG 79 46 0.44 885
544 CCCN{8}CCC 89 53 0.44 886
545 ACAN{12}GCG 23 8 0.43 887
546 AGGN{4}CCG 23 8 0.43 888
547 CGCN{13}GCC 23 8 0.43 889
548 GAGN{2}CGC 23 8 0.42 890
549 CCCN{9}GCG 21 7 0.42 891
550 CGCN{17}ACA 17 5 0.42 892
551 GCGN{17}CCA 23 8 0.42 893
552 AAGN{18}CCG 20 6 0.42 894
553 CGCN{1}GGA 18 5 0.41 895
554 CCAN{1}CCC 90 54 0.41 896
555 CGTN{18}TGC 20 6 0.41 897
556 TCCN{14}CGA 17 5 0.41 898
557 CACN{5}GGG 56 29 0.4 899
558 CCGN{12}GCA 21 7 0.4 900
559 CTGN{6}CCC 77 44 0.4 901
560 CGGN{8}GGC 32 13 0.4 902
561 CCAN{11}GGG 68 38 0.4 903
562 ACGN{19}CAA 21 7 0.39 904
563 GGGN{20}CCC 72 31 0.39 905
564 CGCN{3}CAG 23 8 0.39 906
565 AGCN{17}GGG 58 31 0.37 907
566 CACN{20}CCG 21 7 0.37 908
567 ACGN{17}CAG 24 8 0.37 909
568 AGGN{1}CCC 60 32 0.37 910
569 CGTN{12}CAC 20 6 0.37 911
570 CGGN{9}GGC 23 8 0.37 912
571 CGCN{10}GCG 18 3 0.37 913
572 CCCN{6}CTC 80 47 0.36 914
573 CCGN{10}AGG 23 8 0.36 915
574 CCCN{18}CAG 79 46 0.36 916
575 AGCN{17}CCG 21 7 0.36 917
576 AGCN{9}GCG 18 5 0.36 918
577 CCAN{3}GGC 62 34 0.36 919
578 CCCN{11}GGC 57 30 0.35 920
579 ACGN{5}GCA 23 8 0.35 921
580 CCCN{14}CGG 23 8 0.35 922
581 CCCN{5}CCA 91 55 0.35 923
582 CCGN{1}AGG 22 7 0.34 924
583 GGGN{10}GAC 45 22 0.34 925
584 CGCN{15}CCA 20 6 0.34 926
585 CCTN{19}CGC 22 7 0.34 927
586 CGTN{3}CGC 10 2 0.33 928
587 AGCN{14}CCG 21 7 0.33 929
588 GGCN{2}CGA 17 5 0.33 930
589 CAGN{8}CCC 79 46 0.33 931
590 CCGN{2}GAC 16 4 0.33 932
591 AGCN{19}AGG 70 40 0.32 933
592 CCTN{4}GGC 64 35 0.32 934
593 CCGN{11}AGC 22 7 0.32 935
594 CACN{4}CGC 18 5 0.32 936
595 CCGN{1}CCC 30 12 0.31 937
596 CTGN{13}GGC 73 42 0.31 938
597 CGCN{16}ACC 15 4 0.31 939
598 CACN{18}CAG 79 46 0.31 940
599 GGCN{8}GCC 68 29 0.29 941
600 GGGN{15}GGA 78 46 0.29 942
601 CCGN{16}GCC 22 7 0.29 943
602 CCGN{20}ACC 18 5 0.29 944
603 CGAN{7}CCC 17 5 0.28 945
604 CCGN{6}CTC 23 8 0.28 946
605 CGGN{10}CTC 22 7 0.28 947
606 CAGN{16}CGC 23 8 0.28 948
607 CCAN{3}AGG 77 45 0.27 949
608 GCCN{18}GCC 52 27 0.27 950
609 CGCN{18}GGA 19 6 0.26 951
610 CCGN{20}GGC 22 7 0.26 952
611 ACAN{10}GCG 17 5 0.26 953
612 CGGN{5}CCC 25 9 0.25 954
613 CCCN{7}TCC 75 43 0.25 955
614 ACGN{10}CGC 10 2 0.25 956
615 CCCN{3}TCC 81 48 0.25 957
616 CCGN{8}CGG 20 3 0.24 958
617 CCAN{15}CGG 22 7 0.24 959
618 CCGN{6}CCG 17 5 0.24 960
619 CAGN{3}GCG 25 9 0.24 961
620 GAGN{1}CCC 62 34 0.24 962
621 CCGN{18}TGC 22 7 0.23 963
622 CCCN{7}CCA 85 51 0.23 964
623 CGGN{3}CCA 24 9 0.23 965
624 ACGN{1}CCC 18 5 0.23 966
625 CGGN{13}TGA 21 7 0.22 967
626 CTCN{6}GGC 53 28 0.22 968
627 GCGN{2}GAC 15 4 0.22 969
628 GGGN{11}ACC 49 25 0.22 970
629 CGCN{4}GGA 17 5 0.22 971
630 CCCN{11}CCG 27 10 0.22 972
631 CCGN{19}GCA 20 6 0.22 973
632 GCGN{0}GCA 20 6 0.21 974
633 AGAN{7}CCC 61 33 0.21 975
634 CGGN{2}CCA 21 7 0.21 976
635 CCCN{7}CCC 89 54 0.21 977
636 ACCN{4}GCG 15 4 0.2 978
637 CCTN{15}CGC 20 6 0.2 979
638 AGCN{9}GTC 44 21 0.2 980
639 CCCN{18}CTC 74 43 0.2 981
640 CGCN{18}CGA 9 1 0.19 982
641 CCCN{15}GCC 62 34 0.18 983
642 ACCN{11}GGC 45 22 0.18 984
643 AGGN{15}CGC 29 12 0.18 985
644 GCGN{0}CCA 27 10 0.18 986
645 GCGN{9}AGC 18 5 0.17 987
646 GGGN{18}GCA 59 32 0.17 988
647 CCCN{17}CAG 77 45 0.17 989
648 CCAN{8}CGG 22 8 0.16 990
649 CCGN{10}GGC 21 7 0.16 991
650 GCAN{0}GCC 76 44 0.16 992
651 CAGN{2}CGC 20 6 0.16 993
652 CGCN{8}GGC 19 6 0.16 994
653 CTGN{17}GGC 65 36 0.16 995
654 GGGN{14}ACC 46 23 0.16 996
655 CCGN{1}TGC 20 6 0.16 997
656 CAGN{8}CGC 22 8 0.15 998
657 AAGN{11}CGC 17 5 0.15 999
658 CCGN{6}TCC 22 8 0.14 1000
659 CCAN{18}CCC 72 42 0.14 1001
660 CCAN{0}CCC 84 51 0.14 1002
661 GAGN{6}CCC 53 28 0.14 1003
662 AGCN{20}GGC 52 27 0.14 1004
663 CAGN{0}CGC 21 7 0.14 1005
664 CCGN{12}CTC 22 8 0.14 1006
665 CGCN{15}ACG 9 1 0.13 1007
666 GGCN{17}CGA 15 4 0.13 1008
667 CCGN{16}AAG 19 6 0.13 1009
668 CGCN{14}TCC 19 6 0.12 1010
669 AGGN{7}CGC 20 7 0.12 1011
670 CGGN{7}CCC 22 8 0.12 1012
671 CGCN{4}GCC 34 15 0.12 1013
672 CGAN{6}CCC 17 5 0.12 1014
673 CCCN{19}GGA 60 33 0.11 1015
674 CCCN{16}GCG 28 11 0.11 1016
675 CCAN{7}CGC 20 7 0.11 1017
676 CCCN{6}GCC 80 48 0.11 1018
677 GCCN{14}TCC 55 29 0.11 1019
678 AGGN{14}GCC 64 36 0.1 1020
679 CGCN{11}GCC 20 7 0.1 1021
680 TCCN{0}GCA 17 5 0.09 1022
681 GCGN{8}CCC 27 11 0.09 1023
682 CCAN{11}GCG 19 6 0.09 1024
683 CACN{4}GGG 51 26 0.09 1025
684 CGGN{7}TCC 20 7 0.09 1026
685 GCGN{5}GCC 20 7 0.09 1027
686 ACGN{12}CAG 26 10 0.09 1028
687 CCGN{19}CGC 14 4 0.08 1029
688 CGGN{8}TGC 18 5 0.08 1030
689 CCCN{1}GAG 65 37 0.07 1031
690 GCGN{19}TGA 18 6 0.07 1032
691 GGCN{15}GCC 70 31 0.07 1033
692 CCGN{7}CCC 27 11 0.07 1034
693 ACAN{19}CCC 63 35 0.07 1035
694 ACCN{16}GGG 47 24 0.07 1036
695 AGAN{1}GGC 64 36 0.07 1037
696 GGGN{17}TGA 64 36 0.06 1038
697 CAGN{5}GGG 83 50 0.06 1039
698 GCCN{13}CGC 22 8 0.06 1040
699 GCGN{7}GGA 19 6 0.06 1041
700 CAGN{14}CCA 94 58 0.06 1042
701 CCGN{4}GTC 16 4 0.06 1043
702 CCCN{13}CGC 22 8 0.06 1044
703 GCGN{14}ACC 15 4 0.05 1045
704 CAGN{20}GGG 81 49 0.05 1046
705 CCGN{4}CCC 27 11 0.05 1047
706 CGCN{5}GGC 18 6 0.05 1048
707 CCTN{6}GGC 57 31 0.05 1049
708 AGGN{3}GGC 67 38 0.05 1050
709 CGGN{11}CGC 14 4 0.05 1051
710 CTGN{18}GGA 77 46 0.04 1052
711 CACN{17}CCA 74 43 0.04 1053
712 CGGN{3}GAG 22 8 0.04 1054
713 CCCN{9}CCA 82 49 0.03 1055
714 CCCN{1}ACG 18 6 0.03 1056
715 CAGN{1}GCC 72 42 0.03 1057
716 AGGN{6}CCG 23 8 0.03 1058
717 AGCN{9}GGG 57 31 0.03 1059
718 CCCN{7}GGC 54 29 0.02 1060
719 CCTN{13}CCC 88 54 0.02 1061
720 CCGN{19}TTC 20 7 0.02 1062
721 CCCN{7}CCG 27 11 0.02 1063
722 CGAN{6}GGC 17 5 0.01 1064
723 CGGN{4}CTC 21 7 0.01 1065
724 CGGN{0}CGC 13 3 0.01 1066
725 CCTN{13}ACG 19 6 0.01 1067
726 GGGN{6}CAC 53 28 0.01 1068
727 CCCN{16}CGC 21 7 0.01 1069
728 CCCN{10}CTC 76 45 0 1070
729 CCCN{0}CAG 92 57 0 1071
730 GCCN{5}CCC 65 37 0 1072
TABLE 8
Candidate STAR elements tested by Linear Discriminant Analysis
(SEQ ID NOS: 66-84)
SEQ ID NO: Candidate STAR Location1 Length
66 T2 F 20q13.33 ~2800
67 T2 R 6q14.1 ~2800
68 T3 F 15q12 ~2900
69 T3 R 7q31.2 ~2900
70 T5 F 9q34.13 ND2
71 T5 R 9q34.13 ND
72 T7 22q12.3 ~1200
73 T9 F 21q22.2 ~1600
74 T9 R 22q11.22 ~1600
75 T10 F 7q22.2 ~1300
76 T10 R 6q14.1 ~1300
77 T11 F 17q23.3 ~2000
78 T11 R 16q23.1 ~2000
79 T12 4p15.1 ~2100
80 T13 F 20p13 ~1700
81 T13 R 1p13.3 ~1700
82 T14 R 11q25 ~1500
83 T17 2q31.3 ND
84 T18 2q31.1 ND
1Chromosomal location is determined by BLAT search of DNA sequence data from the STAR elements against the human genome database. The location is given according to standard nomenclature referring to the cytogenetic ideogram of each chromosome; e.g., 1p2.3 is the third cytogenetic sub-band of the second cytogenetic band of the short arm of chromosome 1 (http://www.ncbi.nlm.nih.gov/Class/MLACourse/Genetics/chrombanding.html). F, forward sequencing reaction result; R, reverse sequencing reaction result. When the forward and reverse sequencing results mapped to different genomic locations, each sequence was extended to the full length of the original clone (as determined by restriction mapping) based on sequence information from the human genome database.
2ND: Not Determined.
TABLE 9
Arabidopsis STAR elements of the invention, including
chromosome location and length (SEQ ID NOS: 85-119)
STAR Chromosome Length, kb SEQ ID NO:
A1 I 1.2 85
A2 I 0.9 86
A3 I 0.9 87
A4 I 0.8 88
A5 I 1.3 89
A6 I 1.4 90
A7 II 1.2 91
A8 II 0.8 92
A9 II 0.9 93
A10 II 1.7 94
A11 II 1.9 95
A12 II 1.4 96
A13 II 1.2 97
A14 II 2.1 98
A15 II 1.4 99
A16 II 0.7 100
A17 II 1.5 101
A18 III 1.5 102
A19 III 0.7 103
A20 III 2.0 104
A21 IV 1.8 105
A22 IV 0.8 106
A23 IV 0.6 107
A24 IV 0.5 108
A25 V 0.9 109
A26 V 1.9 110
A27 V 1.1 111
A28 V 1.6 112
A29 V 0.9 113
A30 V 2.0 114
A31 V 2.0 115
A32 V 1.3 116
A33 V 0.9 117
A34 I 0.9 118
A35 II 1.1 119
REFERENCES
  • Altschul S. F., Gish W., Miller W., Myers E. W. and Lipman D. J. (1990) Basic local alignment search tool. J. Mol. Biol. 215, 403-10.
  • Bell A. C., West A. G. and Felsenfeld G. (2001) Insulators and boundaries: versatile regulatory elements in the eukaryotic genome. Science 291, 447-50.
  • Berger J., Hauber J., Hauber R., Geiger R. and Cullen B. R. (1988) Secreted placental alkaline phosphatase: a powerful new quantitative indicator of gene expression in eukaryotic cells. Gene 66, 1-10.
  • Bevan M., Mayer K., White O., Eisen J. A., Preuss D., Bureau T., Salzberg S. L. and Mewes H. W. (2001) Sequence and analysis of the Arabidopsis genome. Curr. Opin. Plant Biol. 4, 105-10.
  • Bibel M. and Barde Y. A. (2000) Neurotrophins: key regulators of cell fate and cell shape in the vertebrate nervous system. Genes Dev. 14, 2919-37.
  • Boivin A. and Dura J. M. (1998) In vivo chromatin accessibility correlates with gene silencing in Drosophila. Genetics 150, 1539-49.
  • Boshart M., Weber F., Jahn G., Dorsch-Hasler K., Fleckenstein B. and Schaffner W. (1985) A very strong enhancer is located upstream of an immediate early gene of human cytomegalovirus. Cell 41, 521-30.
  • Bunker C. A. and Kingston R. E. (1994) Transcriptional repression by Drosophila and mammalian Polycomb group proteins in transfected mammalian cells. Mol. Cell. Biol. 14, 1721-32.
  • Chung J. H, Whiteley M. and Felsenfeld G. (1993) A 5′ element of the chicken beta-globin domain serves as an insulator in human erythroid cells and protects against position effect in Drosophila. Cell 74, 505-14.
  • Deuschle U., Meyer W. K. and Thiesen H. J. (1995) Tetracycline-reversible silencing of eukaryotic promoters. Mol. Cell. Biol. 15, 1907-14.
  • Doll R. F., Crandall J. E., Dyer C. A., Aucoin J. M. and Smith F. I. (1996) Comparison of promoter strengths on gene delivery into mammalian brain cells using AAV vectors. Gene Ther. 3, 437-447.
  • Foecking M. K. and Hofstetter H. (1986) Powerful and versatile enhancer-promoter unit for mammalian expression vectors. Gene 45, 101-5.
  • Garrick D., Fiering S., Martin D. I. and Whitelaw E. (1998) Repeat-induced gene silencing in mammals. Nat. Genet. 18, 56-9.
  • Gerasimova T. I. and Corces V. G. (2001) Chromatin insulators and boundaries: effects on transcription and nuclear organization. Annu. Rev. Genet. 35, 193-208.
  • Gossen M. and Bujard H. (1992) Tight control of gene expression in mammalian cells by tetracycline-responsive promoters. Proc. Natl. Acad. Sci. U.S.A. 89, 5547-51.
  • Graham F. L. and van der Eb A. J. (1973) Transformation of rat cells by DNA of human adenovirus 5. Virology 54, 536-9.
  • Henthorn P., Zervos P., Raducha M., Harris H. and Kadesch T. (1988) Expression of a human placental alkaline phosphatase gene in transfected cells: use as a reporter for studies of gene expression. Proc. Natl. Acad. Sci. U.S.A. 85, 6342-6.
  • Himes S. R. and Shannon M. F. (2000) Assays for transcriptional activity based on the luciferase reporter gene. Methods Mol. Biol. 130, 165-74.
  • Huberty C. J. (1994) Applied discriminant analysis, Wiley and Sons, New York.
  • Initiative A. G. (2000) Analysis of the genome sequence of the flowering plant Arabidopsis thaliana. Nature 408, 796-815.
  • Izumi M. and Gilbert D. M. (1999) Homogeneous tetracycline-regulatable gene expression in mammalian fibroblasts. J. Cell. Biochem. 76, 280-9.
  • Kain S. R. (1997) Use of secreted alkaline phosphatase as a reporter of gene expression in mammalian cells. Methods Mol. Biol. 63, 49-60.
  • Kaufman R. J. (1990) Selection and coamplification of heterologous genes in mammalian cells. Methods in Enzymology 185, 536-566.
  • Kellum R. and Schedl P. (1992) A group of scs elements function as domain boundaries in an enhancer-blocking assay. Mol. Cell. Biol. 12, 2424-2431.
  • Kent W. J. (2002) BLAT—the BLAST-like alignment tool. Genome Res. 12, 656-64.
  • Knofler M., Meinhardt G., Bauer S., Loregger T., Vasicek R., Bloor D. J., Kimber S. J. and Husslein P. (2002) Human Hand1 basic helix-loop-helix (bHLH) protein: extra-embryonic expression pattern, interaction partners and identification of its transcriptional repressor domains. Biochem J. 361, 641-51.
  • Meyer P. (2000) Transcriptional transgene silencing and chromatin components. Plant Mol. Biol. 43, 221-34.
  • Mercenier A., Wiedermann U. and Breiteneder H. (2001) Edible genetically modified microorganisms and plants for improved health. Curr. Opin. Biotechnol. 12, 510-5.
  • Morgenstern J. P. and Land H. (1990) Advanced mammalian gene transfer: high titre retroviral vectors with multiple drug selection markers and a complementary helper-free packaging cell line. Nucleic Acids Res. 18, 3587-96.
  • Sambrook J., Fritsch E. F. and Maniatis T. (1989) Molecular Cloning: A Laboratory Manual, Second ed., Cold Spring Harbor Laboratory Press, Plainview N.Y.
  • Sanger F., Nicklen S, and Coulson A. R. (1977) DNA sequencing with chain-terminating inhibitors. Proc. Natl. Acad. Sci. U.S.A. 74, 5463-7.
  • Stam M., Viterbo A., Mol. J. N. and Kooter J. M. (1998) Position-dependent methylation and transcriptional silencing of transgenes in inverted T-DNA repeats: implications for posttranscriptional silencing of homologous host genes in plants. Mol. Cell. Biol. 18, 6165-77.
  • Umana P., Jean-Mairet J. and Bailey J. E. (1999) Tetracycline-regulated overexpression of glycosyltransferases in Chinese hamster ovary cells. Biotechnol. Bioeng. 65, 542-9.
  • Van der Vlag J., den Blaauwen J. L., Sewalt R. G., van Driel R. and Otte A. P. (2000) Transcriptional repression mediated by polycomb group proteins and other chromatin-associated repressors is selectively blocked by insulators. J. Biol. Chem. 275, 697-704.
  • van Helden J., Andre B. and Collado-Vides J. (1998) Extracting regulatory sites from the upstream region of yeast genes by computational analysis of oligonucleotide frequencies J. Mol. Biol. 281, 827-42.
  • van Helden J., Andre B. and Collado-Vides J. (2000) A web site for the computational analysis of yeast regulatory sequences. Yeast 16, 177-87.
  • van Helden J., Rios A. F. and Collado-Vides J. (2000) Discovering regulatory elements in non-coding sequences by analysis of spaced dyads. Nucleic Acids Res. 28, 1808-18.
  • Vance V. and Vaucheret H. (2001) RNA silencing in plants—defense and counterdefense. Science 292, 2277-80.
  • Wigler M., Pellicer A., Silverstein S, and Axel R. (1978) Biochemical transfer of single-copy eucaryotic genes using total cellular DNA as donor. Cell 14, 725-31.
  • Yang T. T., Sinai P., Kitts P. A. and Kain S. R. (1997) Quantification of gene expression with a secreted alkaline phosphatase reporter system. Biotechniques 23, 1110-4.
  • Zink D. and Paro R. (1995) Drosophila Polycomb-group regulated chromatin inhibits the accessibility of a trans-activator to its target DNA. Embo. J. 14, 5660-71.

Claims (18)

1. A method for producing a proteinaceous molecule in a cell comprising:
providing a cell selected from the group consisting of: a cell having an adenovirus Early Region 1 (E1) sequence, a HuNS-1 myeloma cell, a 293 cell, a CHO cell, a Vero cell, a WERI-Rb-1 retinoblastoma cell, a BHK cell, a non-secreting mouse myeloma Sp2/0-Ag 14 cell, a non-secreting mouse myeloma NSO cell, and an NCI-H295R adrenal gland carcinoma cell;
wherein said cell comprises an anti-repressor activity sequence operably linked to a nucleic acid sequence encoding a heterologous proteinaceous molecule, wherein said anti-repressor activity sequence comprises SEQ ID NO:44;
expressing the proteinaceous molecule in said cell; and
isolating said proteinaceous molecule.
2. The method of claim 1, wherein said anti-repressor activity sequence is SEQ ID NO:44.
3. The method of claim 1, wherein said cell comprises an adenovirus Early Region 1 (E1) sequence.
4. The method of claim 1, wherein said proteinaceous molecule is secreted by said cell.
5. The method of claim 2, wherein said proteinaceous molecule is secreted by said cell.
6. The method of claim 1, wherein said cell comprises a plurality of the anti-repressor activity sequence operably linked to said nucleic acid sequence encoding the heterologous proteinaceous molecule.
7. The method of claim 6, wherein at least one anti-repressor activity sequence is positioned 5′ of the sequence encoding the proteinaceous molecule and at least one anti-repressor activity sequence is positioned 3′ of the sequence encoding the proteinaceous molecule.
8. The method of claim 1, wherein said cell is a CHO cell.
9. The method of claim 6, wherein said cell is a CHO cell.
10. The method of claim 7, wherein said cell is a CHO cell.
11. A recombinant host cell line, comprising:
a cell selected from the group consisting of: a cell line comprising an adenovirus Early Region 1 (E1) sequence, a HuNS-1 myeloma cell line, a 293 cell line, a CHO cell line, a Vero cell line, a WERI-Rb-1 retinoblastoma cell line, a BHK cell line, a non-secreting mouse myeloma Sp2/0-Ag 14 cell line, a non-secreting mouse myeloma NSO cell line, and an NCI-H295R adrenal gland carcinoma cell line;
said cell comprising an anti-repressor activity sequence operably linked to a nucleic acid sequence encoding a heterologous proteinaceous molecule, wherein said anti-repressor activity sequence comprises SEQ ID NO:44.
12. The cell line of claim 11, wherein said anti-repressor activity sequence is SEQ ID NO:44.
13. The cell line of claim 11, wherein said cell line comprises an adenovirus Early Region 1 sequence.
14. The cell line of claim 11, wherein said cell comprises a plurality of the anti-repressor activity sequence operably linked to said nucleic acid sequence encoding the heterologous proteinaceous molecule.
15. The cell line of claim 11, wherein at least one anti-repressor activity sequence is positioned 5′ of the sequence encoding the proteinaceous molecule and at least one anti-repressor activity sequence is positioned 3′ of the sequence encoding the proteinaceous molecule.
16. The cell line of claim 11, wherein said cell line is a CHO cell line.
17. The cell line of claim 14, wherein said cell line is a CHO cell line.
18. The cell line of claim 15, wherein said cell line is a CHO cell line.
US11/888,568 2002-06-14 2007-07-31 Means and methods for regulating gene expression Expired - Fee Related US7794977B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/888,568 US7794977B2 (en) 2002-06-14 2007-07-31 Means and methods for regulating gene expression
US11/978,483 US20080206813A1 (en) 2002-06-14 2007-10-26 Means and methods for regulating gene expression

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
EP02077344 2002-06-14
EP02077344.6 2002-06-14
EP02077344 2002-06-14
PCT/NL2003/000410 WO2003106674A2 (en) 2002-06-14 2003-05-30 Means and methods for regulating gene expression
US11/012,546 US7267965B2 (en) 2002-06-14 2004-12-14 Means and methods for regulating gene expression
US11/888,568 US7794977B2 (en) 2002-06-14 2007-07-31 Means and methods for regulating gene expression

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/012,546 Division US7267965B2 (en) 2002-06-14 2004-12-14 Means and methods for regulating gene expression

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/978,483 Division US20080206813A1 (en) 2002-06-14 2007-10-26 Means and methods for regulating gene expression

Publications (2)

Publication Number Publication Date
US20080131930A1 US20080131930A1 (en) 2008-06-05
US7794977B2 true US7794977B2 (en) 2010-09-14

Family

ID=29724493

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/012,546 Expired - Fee Related US7267965B2 (en) 2002-06-14 2004-12-14 Means and methods for regulating gene expression
US11/888,568 Expired - Fee Related US7794977B2 (en) 2002-06-14 2007-07-31 Means and methods for regulating gene expression
US11/978,483 Abandoned US20080206813A1 (en) 2002-06-14 2007-10-26 Means and methods for regulating gene expression

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/012,546 Expired - Fee Related US7267965B2 (en) 2002-06-14 2004-12-14 Means and methods for regulating gene expression

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/978,483 Abandoned US20080206813A1 (en) 2002-06-14 2007-10-26 Means and methods for regulating gene expression

Country Status (4)

Country Link
US (3) US7267965B2 (en)
EP (1) EP1513936A2 (en)
AU (1) AU2003243059A1 (en)
WO (1) WO2003106674A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060172382A1 (en) * 2004-11-08 2006-08-03 Chromagenics B.V. Selection of host cells expressing protein at high levels
US9228004B2 (en) 2004-11-08 2016-01-05 Chromagenics B.V. Selection of host cells expressing protein at high levels

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MXPA03011801A (en) * 2001-07-04 2005-03-07 Chromagenics Bv Method of selecting dna sequence with transcription modulating activity using a vector comprising an element with a gene transcription repressing activity.
WO2003106674A2 (en) * 2002-06-14 2003-12-24 Chromagenics B.V. Means and methods for regulating gene expression
JP4647309B2 (en) * 2002-06-14 2011-03-09 クロマジェニックス ビー.ブイ. Methods for simultaneous production of multiple proteins; vectors and cells for use therein
USRE47770E1 (en) 2002-07-18 2019-12-17 Merus N.V. Recombinant production of mixtures of antibodies
EP1523496B1 (en) 2002-07-18 2011-06-29 Merus B.V. Recombinant production of mixtures of antibodies
AU2003290453A1 (en) * 2002-12-20 2004-07-14 Chromagenics B.V. Means and methods for producing a protein through chromatin openers that are capable of rendering chromatin more accessible to transcription factors
WO2004106375A1 (en) 2003-05-30 2004-12-09 Merus Biopharmaceuticals B.V. I.O. Fab library for the preparation of anti vegf and anti rabies virus fabs
US20100069614A1 (en) 2008-06-27 2010-03-18 Merus B.V. Antibody producing non-human mammals
US8999667B2 (en) * 2004-11-08 2015-04-07 Chromagenics B.V. Selection of host cells expressing protein at high levels
US20100136616A1 (en) * 2004-11-08 2010-06-03 Chromagenics B.V. Selection of Host Cells Expressing Protein at High Levels
CA2581422C (en) * 2004-11-08 2012-05-22 Chromagenics B.V. Selection of host cells expressing protein at high levels
CN100386437C (en) * 2006-05-10 2008-05-07 扬州大学 Method for constructing idiosyncratic carrier of galactophore of transgenic animal in high expression level
CN102459607B (en) 2009-06-15 2014-08-06 萨拉基尼克有限公司 Novel stringent selectable markers
EP2611915B1 (en) 2010-09-01 2015-04-15 Cellagenics B.V. Nucleic acid fragments from a ribosomal protein promoter for enhancing gene expression
SG10201607371UA (en) 2012-04-20 2016-10-28 Merus Nv Methods and means for the production of ig-like molecules

Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996004390A1 (en) 1994-07-29 1996-02-15 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Dna sequence which acts as a chromatin insulator element to protect expressed genes from cis-acting regulatory sequences in mammalian cells
WO1997027207A1 (en) 1996-01-26 1997-07-31 North Carolina State University A plant nuclear scaffold attachment region which increases gene expression
WO1998011207A2 (en) 1996-09-16 1998-03-19 Geron Corporation Assays for regulators of mammalian telomerase expression
WO1998049289A1 (en) 1997-05-01 1998-11-05 Icos Corporation HAMSTER EF-1α TRANSCRIPTIONAL REGULATORY DNA
WO2000005393A2 (en) 1998-07-21 2000-02-03 Cobra Therapeutics Limited A polynucleotide comprising a ubiquitous chromatin opening element (ucoe)
WO2000009749A1 (en) 1998-08-14 2000-02-24 Stichting Voor De Technische Wetenschappen Method of detecting a dna sequence, a dna sequence, a method of making a dna construct and the use thereof
WO2000017337A1 (en) 1998-09-22 2000-03-30 Bristol-Myers Squibb Company Expression vectors containing hot spot for increased recombinant protein expression in transfected cells
WO2000023606A1 (en) 1998-10-22 2000-04-27 Medical College Of Georgia Institute, Inc. Long terminal repeat, enhancer, and insulator sequences for use in recombinant vectors
WO2001059118A1 (en) 2000-02-11 2001-08-16 Millennium Pharmaceuticals, Inc. 43239, a gpcr-like molecule and uses thereof
WO2001059117A2 (en) 2000-02-11 2001-08-16 Millennium Pharmaceuticals, Inc. Seven-transmembrane proteins/g-protein coupled receptors
WO2002024930A2 (en) 2000-09-20 2002-03-28 Ml Laboratories Plc Artificial ubiquitous chromatin opening elements (ucoe)
EP1273666A1 (en) 2001-07-04 2003-01-08 Chromagenics B.V. Method of selecting a DNA sequence with transcription modulating activity using a vector comprising an element with a gene transcription repressing activity
WO2003004704A2 (en) 2001-07-04 2003-01-16 Chromagenics B.V. Method of selecting dna sequence with transcription modulating activity using a vector comprising an element with a gene transcription repressing activity
WO2004055215A1 (en) 2002-12-18 2004-07-01 Chromagenics B.V. A method for improving protein production
WO2004056986A2 (en) 2002-12-20 2004-07-08 Chromagenics B.V. Means and methods for producing a protein through chromatin openers that are capable of rendering chromatin more accessible to transcription factors
US6800457B2 (en) 1998-09-22 2004-10-05 Bristol-Myers Squibb Company Expression vectors containing hot spot for increased recombinant protein expression in transfected cells
US7267965B2 (en) 2002-06-14 2007-09-11 Chromagenics B.V. Means and methods for regulating gene expression

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4647309B2 (en) * 2002-06-14 2011-03-09 クロマジェニックス ビー.ブイ. Methods for simultaneous production of multiple proteins; vectors and cells for use therein

Patent Citations (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5610053A (en) 1993-04-07 1997-03-11 The United States Of America As Represented By The Department Of Health And Human Services DNA sequence which acts as a chromatin insulator element to protect expressed genes from cis-acting regulatory sequences in mammalian cells
WO1996004390A1 (en) 1994-07-29 1996-02-15 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Dna sequence which acts as a chromatin insulator element to protect expressed genes from cis-acting regulatory sequences in mammalian cells
WO1997027207A1 (en) 1996-01-26 1997-07-31 North Carolina State University A plant nuclear scaffold attachment region which increases gene expression
US5773695A (en) 1996-01-26 1998-06-30 North Carolina State University Plant nuclear scaffold attachment region and method for increasing gene expression in transgenic cells
WO1998011207A2 (en) 1996-09-16 1998-03-19 Geron Corporation Assays for regulators of mammalian telomerase expression
WO1998049289A1 (en) 1997-05-01 1998-11-05 Icos Corporation HAMSTER EF-1α TRANSCRIPTIONAL REGULATORY DNA
US5888809A (en) 1997-05-01 1999-03-30 Icos Corporation Hamster EF-1α transcriptional regulatory DNA
WO2000005393A2 (en) 1998-07-21 2000-02-03 Cobra Therapeutics Limited A polynucleotide comprising a ubiquitous chromatin opening element (ucoe)
US20050106609A1 (en) 1998-08-14 2005-05-19 Arie Otte Method of detecting a DNA sequence, a DNA sequence, a method of making a DNA construct and the use thereof
WO2000009749A1 (en) 1998-08-14 2000-02-24 Stichting Voor De Technische Wetenschappen Method of detecting a dna sequence, a dna sequence, a method of making a dna construct and the use thereof
US6872524B1 (en) 1998-08-14 2005-03-29 Chromagenics B.V. Method of detecting an expression-enhancing sequence
WO2000017337A1 (en) 1998-09-22 2000-03-30 Bristol-Myers Squibb Company Expression vectors containing hot spot for increased recombinant protein expression in transfected cells
US6800457B2 (en) 1998-09-22 2004-10-05 Bristol-Myers Squibb Company Expression vectors containing hot spot for increased recombinant protein expression in transfected cells
US6521419B1 (en) 1998-09-22 2003-02-18 Kanakaraju Koduri Expression vectors containing hot spot for increased recombinant protein expression in transfected cells
WO2000023606A1 (en) 1998-10-22 2000-04-27 Medical College Of Georgia Institute, Inc. Long terminal repeat, enhancer, and insulator sequences for use in recombinant vectors
US6395549B1 (en) 1998-10-22 2002-05-28 Medical College Of Georgia Research Institute, Inc. Long terminal repeat, enhancer, and insulator sequences for use in recombinant vectors
WO2001059118A1 (en) 2000-02-11 2001-08-16 Millennium Pharmaceuticals, Inc. 43239, a gpcr-like molecule and uses thereof
WO2001059117A2 (en) 2000-02-11 2001-08-16 Millennium Pharmaceuticals, Inc. Seven-transmembrane proteins/g-protein coupled receptors
US6586205B1 (en) 2000-02-11 2003-07-01 Millennium Pharmaceuticals, Inc. 43239 a novel GPCR-like molecule and uses thereof
WO2002024930A2 (en) 2000-09-20 2002-03-28 Ml Laboratories Plc Artificial ubiquitous chromatin opening elements (ucoe)
WO2003004704A2 (en) 2001-07-04 2003-01-16 Chromagenics B.V. Method of selecting dna sequence with transcription modulating activity using a vector comprising an element with a gene transcription repressing activity
EP1273666A1 (en) 2001-07-04 2003-01-08 Chromagenics B.V. Method of selecting a DNA sequence with transcription modulating activity using a vector comprising an element with a gene transcription repressing activity
US20070026499A1 (en) * 2001-07-04 2007-02-01 Chromagenics B.V. Nucleic acid sequences having gene transcription regulatory qualities
US20070026498A1 (en) * 2001-07-04 2007-02-01 Chromagenics B.V. Nucleic acid sequences having gene transcription regulatory qualities
US20070031935A1 (en) * 2001-07-04 2007-02-08 Chromagenics B.V. Nucleic acid sequences having gene transcription regulatory qualities
US20070031933A1 (en) * 2001-07-04 2007-02-08 Chromagenics B.V. Nucleic acid sequences having gene transcription regulatory qualities
US20070031934A1 (en) * 2001-07-04 2007-02-08 Chromagenics B.V. Nucleic acid sequences having gene transcription regulatory qualities
US20070037256A1 (en) * 2001-07-04 2007-02-15 Chromagenics B.V. Nucleic acid sequences having gene transcription regulatory qualities
US7192741B2 (en) * 2001-07-04 2007-03-20 Chromagenics B.V. DNA sequences comprising gene transcription regulatory qualities and methods for detecting and using such DNA sequences
US20070128717A1 (en) * 2001-07-04 2007-06-07 Chromagenics B.V. DNA sequences comprising gene transcription regulatory qualities and methods for detecting and using such DNA sequences
US7267965B2 (en) 2002-06-14 2007-09-11 Chromagenics B.V. Means and methods for regulating gene expression
WO2004055215A1 (en) 2002-12-18 2004-07-01 Chromagenics B.V. A method for improving protein production
WO2004056986A2 (en) 2002-12-20 2004-07-08 Chromagenics B.V. Means and methods for producing a protein through chromatin openers that are capable of rendering chromatin more accessible to transcription factors

Non-Patent Citations (64)

* Cited by examiner, † Cited by third party
Title
Aranda et al., Definition of Transcriptional Pause Elements in Fission Yeast, Molecular and Cellular Biology, Feb. 1999, pp. 1251-61, vol. 19, No. 2.
Auten et al., "Effect of Scaffold Attachment Region on Transgene Expression in Retrovirus Vector-Transduced Primary T cells and Macrophages," Human Gene Therapy, May 20, 1999, pp. 1389-1399, vol. 10, No. 8.
Bell et al., Insulators and Boundaries: Versatile Regulatory Elements in the Eukaryotic Genome, Science, pp. 447-450, vol. 291, No. 5503.
Burgess-Beusse et al., The insulation of genes from external enhancers and silencing chromatin, PNAS, Dec. 10, 2002, pp. 16433-37, vol. 99, Suppl. 4.
Chan et al., p300-CBP proteins: HATs for transcriptional bridges and scaffolds, Journal of Cell Science, 2001, pp. 2363-2373, vol. 114.
Chung et al., A 5' Element of the Chicken Beta-Globin Domain Serves as an Insulator in Human Erythroid Cells and Protects against Position Effect in Drosophila, Aug. 13, 1993, Cell, pp. 505-514, vol. 74.
Chung et al., A 5′ Element of the Chicken Beta-Globin Domain Serves as an Insulator in Human Erythroid Cells and Protects against Position Effect in Drosophila, Aug. 13, 1993, Cell, pp. 505-514, vol. 74.
Database EMBL ′Online! Apr. 26, 2001, "RST28606 Athersys RAGE Library Homo sapiens cDNA, mRNA sequence," XP002359990 retrieved from EBI accession No. EM—PRO:BG209092, database accession No. BG209092 for SEQ ID No. 40.
Database EMBL ′Online! Aug. 4, 1999, "Homo sapiens chromosome 19 clone CTD-2540B15, complete sequence," XP002359985 retrieved from EBI accession No. EM—PRO:AC008738, database accession No. AC008738 for SEQ ID No. 7.
Database EMBL ′Online! Dec. 15, 1999, "Homo sapiens BAC clone RP11-572N21 from 2, complete sequence," XP002359988 retrieved from EBI accession No. EM—PRO:AC018470, database accession No. AC018470, for SEQ ID No. 17.
Database EMBL ′Online! Dec. 23, 1999, "Human DNA sequence from clone RP11-54H19 on chromosome 1 Contains the 3′ end of the LMNA gene for lamin A/C, the gene for a novel protein similar to semaphorins (FLJ12287), a novel gene (KIAA0446), the PMFI gene for polyamine-modulated factor 1, the BGLAP gene for bone gamma-carboxyglutamate (gla) p," XP002359989, retrieved from EBI accession No. EM—PRO:AL135927, database accession No. AL135927 for SEQ ID No. 27.
Database EMBL ′Online! Feb. 3, 2004, "Sequence 33099 from Patent W002068579," XP002359986 retrieved from EBI accession No. EM—PRO:CQ747165, database accession No. CQ747165 for SEQ ID No. 9.
Database EMBL ′Online! Jan. 25, 2001, "QV2-NN0045-081200-535-c10 NN0045 Homo sapiens cDNA, mRNA sequence." XP002359993 retrieved from EBI accession No. EM—PRO:BF960930, database accession No. BF960930 for SEQ ID No. 43.
Database EMBL ′Online! Mar. 15, 1999, "Homo sapiens chromosome UNK clone CTA-435J10, working draft sequence, 1 unordered pieces," XP002359997 retrieved from EBI accession No. EM—PRO:AC007044, database accession No. AC007044 for SEQ ID No. 61.
Database EMBL ′Online! Mar. 19, 1998, CIT-HSP-2172C8.TF CIT-HSP Homo sapiens genomic clone 2172C8, genomic survey sequence, XP002359995 retrieved from EBI accession No. EM—PRO:B92131, database accession No. B92131 for SEQ ID No. 44.
Database EMBL ′Online! Oct. 28, 1998, "Homo sapiens neurexin III-alpha gene, partial cds," XP002359992, retrieved from EBI accession No. EM—PRO:AF099810, database accession No. AF099810 for SEQ ID No. 43.
Database EMBL ′Online! Sep. 24, 2000, "Homo sapiens chromosome 4 clone RP11-680II8, working draft sequence, 25 unordered pieces," XP002359987 retrieved from EBI accession No. EM—PRO:AC080087, database accession No. AC080087 for SEQ ID No. 9.
Database EMBL ′Online! Sep. 29, 1999, "Homo sapiens genomic DNA, chromosome 22q11.2, Cat Eye Syndrome region, clone:c91G6," XP002359996 retrieved from EBI accession No. EM—PRO:AP000528, database accession No. AP000528 for SEQ ID No. 45.
Database EMBL ′Online! Sep. 29, 1999, "Homo sapiens genomic DNA, chromosome 22q11.2, Cat Eye Syndrome region, clone:N64E9," XP002359991, retrieved from EBI accession No. EM—PRO:AP000526, database accession No. AP000526 for SEQ ID No. 40.
Database EMBL ′Online! Sep. 29, 1999, Homo sapiens genomic DNA, chromosome 22q11.2, Cat Eye Syndrome region, clone:N14H11, XP002359994 retrieved from EBI accession No. EM—PRO:AP000525, database accession No. AP000525 for SEQ ID No. 44.
Database EMBL ′Online!, Jul. 8, 1992, H. sapiens HOX4B gene upstream sequence XP002348163 retrieved from EBI, Database accession No. X67079, Abstract.
Database EMBL 'Online! Apr. 26, 2001, "RST28606 Athersys RAGE Library Homo sapiens cDNA, mRNA sequence," XP002359990 retrieved from EBI accession No. EM-PRO:BG209092, database accession No. BG209092 for SEQ ID No. 40.
Database EMBL 'Online! Aug. 4, 1999, "Homo sapiens chromosome 19 clone CTD-2540B15, complete sequence," XP002359985 retrieved from EBI accession No. EM-PRO:AC008738, database accession No. AC008738 for SEQ ID No. 7.
Database EMBL 'Online! Dec. 15, 1999, "Homo sapiens BAC clone RP11-572N21 from 2, complete sequence," XP002359988 retrieved from EBI accession No. EM-PRO:AC018470, database accession No. AC018470, for SEQ ID No. 17.
Database EMBL 'Online! Dec. 23, 1999, "Human DNA sequence from clone RP11-54H19 on chromosome 1 Contains the 3' end of the LMNA gene for lamin A/C, the gene for a novel protein similar to semaphorins (FLJ12287), a novel gene (KIAA0446), the PMFI gene for polyamine-modulated factor 1, the BGLAP gene for bone gamma-carboxyglutamate (gla) p," XP002359989, retrieved from EBI accession No. EM-PRO:AL135927, database accession No. AL135927 for SEQ ID No. 27.
Database EMBL 'Online! Feb. 3, 2004, "Sequence 33099 from Patent W002068579," XP002359986 retrieved from EBI accession No. EM-PRO:CQ747165, database accession No. CQ747165 for SEQ ID No. 9.
Database EMBL 'Online! Jan. 25, 2001, "QV2-NN0045-081200-535-c10 NN0045 Homo sapiens cDNA, mRNA sequence." XP002359993 retrieved from EBI accession No. EM-PRO:BF960930, database accession No. BF960930 for SEQ ID No. 43.
Database EMBL 'Online! Mar. 15, 1999, "Homo sapiens chromosome UNK clone CTA-435J10, working draft sequence, 1 unordered pieces," XP002359997 retrieved from EBI accession No. EM-PRO:AC007044, database accession No. AC007044 for SEQ ID No. 61.
Database EMBL 'Online! Mar. 19, 1998, CIT-HSP-2172C8.TF CIT-HSP Homo sapiens genomic clone 2172C8, genomic survey sequence, XP002359995 retrieved from EBI accession No. EM-PRO:B92131, database accession No. B92131 for SEQ ID No. 44.
Database EMBL 'Online! Oct. 28, 1998, "Homo sapiens neurexin III-alpha gene, partial cds," XP002359992, retrieved from EBI accession No. EM-PRO:AF099810, database accession No. AF099810 for SEQ ID No. 43.
Database EMBL 'Online! Sep. 24, 2000, "Homo sapiens chromosome 4 clone RP11-680II8, working draft sequence, 25 unordered pieces," XP002359987 retrieved from EBI accession No. EM-PRO:AC080087, database accession No. AC080087 for SEQ ID No. 9.
Database EMBL 'Online! Sep. 29, 1999, "Homo sapiens genomic DNA, chromosome 22q11.2, Cat Eye Syndrome region, clone:c91G6," XP002359996 retrieved from EBI accession No. EM-PRO:AP000528, database accession No. AP000528 for SEQ ID No. 45.
Database EMBL 'Online! Sep. 29, 1999, "Homo sapiens genomic DNA, chromosome 22q11.2, Cat Eye Syndrome region, clone:N64E9," XP002359991, retrieved from EBI accession No. EM-PRO:AP000526, database accession No. AP000526 for SEQ ID No. 40.
Database EMBL 'Online! Sep. 29, 1999, Homo sapiens genomic DNA, chromosome 22q11.2, Cat Eye Syndrome region, clone:N14H11, XP002359994 retrieved from EBI accession No. EM-PRO:AP000525, database accession No. AP000525 for SEQ ID No. 44.
Database EMBL 'Online!, Jul. 8, 1992, H. sapiens HOX4B gene upstream sequence XP002348163 retrieved from EBI, Database accession No. X67079, Abstract.
Eggermont et al., Poly(A) signals and transcriptional pause sites combine to prevent interference between RNA polymerase II promoters, The EMBO Journal, 1993, pp. 2539-2548, vol. 12, No. 6.
Emery et al., "A chromatin insulator protects retrovirus vectors from chromosomal position effects," Proceedings of the National Academy of Sciences of USA, Aug. 1, 2000, pp. 9150-9155, vol. 97, No. 16.
European Search Report dated Dec. 22, 2005.
Farrell et al., Conserved CTCF Insulator Elements Flank the Mouse and Human Beta-Globin Loci, Molecular and Cellular Biology, Jun. 2002, pp. 3820-3831, vol. 22. No. 11.
Glucksmann et al., Database accession No. AAH76193, Oct. 29, 2001.
Han et al., "Matrix attachment regions (MARs) enhance transformation frequency and transgene expression in poplar," Transgenic Research, 1997, pp. 415-420, vol. 6.
Johnson et al., Requirements for utilization of CREB binding protein by hypersensitive site two of the Beta-globin locus control region, Nucleic Acids Research 2002, pp. 1522-1530, vol. 30, No. 7.
Kellum et al., A Group of scs Elements Function as Domain Boundaries in an Enhancer-Blocking Assay, Molecular and Cellular Biology, May 1992, pp. 2424-2431, vol. 12, No. 5.
Kwaks et al., "Identification of anti-repressor elements that confer high stable protein in production in mammalian cells," Nature Biotechnology, May 20, 2003, pp. 553-558, vol. 21, No. 5.
Maniatis et al., Recognition Sequences of Repressor and Polymerase in the Operators of Bacteriophage Lambda, Cell, Jun. 1975, pp. 109-113, vol. 5.
Martinez-Balbas et al., The acetyltransferase activity of CBP stimulates transcription, The EMBO Journal, 1998, pp. 2886-2893, vol. 17, No. 10.
Mielke et al., "Stabilized, long-term expression of heterodimeric proteins from tricistronic mRNA," Gene, Aug. 22, 2000, pp. 1-8, vol. 254, No. 1-2.
Migliaccio et al., "Stable and unstable transgene integration sites in the human genome: extinction of the Green Fluorescent Protein transgene in K562 cells," Gene, Oct. 3, 2000, pp. 197-214, vol. 256, No. 1-2.
Partial European Search Report, EP 05 07 6209, dated Oct. 7, 2005.
PCT International Search Report, PCT/NL2003/00410, dated Dec. 15, 2003.
Pile et al., "GAGA Factor-dependent Transcription and Establishment of DNase Hypersensitivity Are Independent and Unrelated Events In Vivo," J. of Biological Chemistry, Jan. 14, 2000, pp. 1398-1404, vol. 275, No. 2.
Reik et al., Biotechnologies and therapeutics: Chromatin as a target, Current Opinion in Genetics & Development, 2002, pp. 233-242, vol. 12.
Seum et al., A GAL4-HP1 fusion protein targeted near heterochromatin promotes gene silencing, Chromosoma, 2000, pp. 453-459, vol. 109.
Sigrist et al., "Chromatin Insulator Elements Black the Silencing of a Target Gene by the Drosophila Polycomb Response Element (PRE) but Allow trans Interactions Between PREs on Different Chromosomes," Genetics, Sep. 1997, pp. 209-211, vol. 147, No. 1.
U.S. Appl. No. 11/899,505, filed Sep. 6, 2007, Otte et al., Selection of Host Cells Expressing Protein at High Levels.
U.S. Appl. No. 11/978,109, filed Oct. 26, 2007, Otte et al., A Method for Simultaneous Production of Multiple Proteins; Vectors and Cells for use Therein.
U.S. Appl. No. 11/978,134, filed Oct. 26, 2007, Otte et al., A Method for Simultaneous Production of Multiple Proteins; Vectors and Cells for use Therein.
U.S. Appl. No. 11/978,483, filed Oct. 26, 2007, Otte et al., Means and Methods for Regulating Gene Expression.
U.S. Appl. No. 12/218,128, filed Jul. 11, 2008, Otte et al., A Method for Improving Protein Production.
U.S. Appl. No. 12/223,801, filed Aug. 6, 2008, Otte et al., Selection of Host Cells Expressing Protein at High Levels.
U.S. Appl. No. 12/225,355, filed Sep. 19, 2008, Otte et al., Expression Augmenting DNA Fragments, use Thereof, and Methods for Finding Thereof.
U.S. Appl. No. 12/226,706, filed Oct. 24, 2008, Otte et al., Selection of Host Cells Expressing Protein at High Levels.
Van Der Vlag et al., "Transcriptional Repression Mediated by Polycomb Group Proteins and Other Chromatin-associated Repressors Is Selectively Blacked by Insulators," J. of Biological Chemistry, Jan. 7, 2000, pp. 697-704, vol. 275, No. 1.
West et al., "Insulators: many functions, many mechanisms," Genes and Development, 1 Feb. 2002, pp. 271-288, vol. 16, No. 3.

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060172382A1 (en) * 2004-11-08 2006-08-03 Chromagenics B.V. Selection of host cells expressing protein at high levels
US20110014655A1 (en) * 2004-11-08 2011-01-20 Chromagenics B.V. Selection of host cells expressing protein at high levels
US8771984B2 (en) 2004-11-08 2014-07-08 Chromagenics B.V. Selection of host cells expressing protein at high levels
US9228004B2 (en) 2004-11-08 2016-01-05 Chromagenics B.V. Selection of host cells expressing protein at high levels

Also Published As

Publication number Publication date
AU2003243059A8 (en) 2003-12-31
US20080131930A1 (en) 2008-06-05
WO2003106674A2 (en) 2003-12-24
AU2003243059A1 (en) 2003-12-31
US20050214906A1 (en) 2005-09-29
US20080206813A1 (en) 2008-08-28
WO2003106674A3 (en) 2004-03-04
US7267965B2 (en) 2007-09-11
EP1513936A2 (en) 2005-03-16

Similar Documents

Publication Publication Date Title
US7794977B2 (en) Means and methods for regulating gene expression
US7960143B2 (en) Method for simultaneous production of multiple proteins; vectors and cells for use therein
US7192741B2 (en) DNA sequences comprising gene transcription regulatory qualities and methods for detecting and using such DNA sequences
CA2812821C (en) Dna sequences comprising gene transcription regulatory qualities and methods for detecting and using such dna sequences
AU2008202251B2 (en) A method for simultaneous production of multiple proteins; vectors and cells for use therein
AU2011218621B2 (en) A method for simultaneous production of multiple proteins; vectors and cells for use therein
Class et al. Patent application title: Method for simultaneous production of multiple proteins; vectors and cells for use therein Inventors: Arie Pieter Otte (Amersfoort, NL) Arie Pieter Otte (Amersfoort, NL) Arthur Leo Kruckeberg (Shoreline, WA, US) Richard George Antonius Bernardus Sewalt (Arnhem, NL) Assignees: Crucell Holland BV

Legal Events

Date Code Title Description
AS Assignment

Owner name: CHROMAGENICS B.V., NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:OTTE, ARIE P.;KRUCKEBERG, ARTHUR L.;SATIJN, DAVID P. E.;REEL/FRAME:019705/0283;SIGNING DATES FROM 20050418 TO 20050525

Owner name: CHROMAGENICS B.V., NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:OTTE, ARIE P.;KRUCKEBERG, ARTHUR L.;SATIJN, DAVID P. E.;SIGNING DATES FROM 20050418 TO 20050525;REEL/FRAME:019705/0283

STCF Information on status: patent grant

Free format text: PATENTED CASE

FPAY Fee payment

Year of fee payment: 4

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552)

Year of fee payment: 8

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20220914